US11920174B2 - RNA analysis by total hydrolysis and quantification of released nucleosides - Google Patents

RNA analysis by total hydrolysis and quantification of released nucleosides Download PDF

Info

Publication number
US11920174B2
US11920174B2 US16/081,863 US201716081863A US11920174B2 US 11920174 B2 US11920174 B2 US 11920174B2 US 201716081863 A US201716081863 A US 201716081863A US 11920174 B2 US11920174 B2 US 11920174B2
Authority
US
United States
Prior art keywords
rna molecules
rna
sample
length
molecules
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US16/081,863
Other languages
English (en)
Other versions
US20190100784A1 (en
Inventor
Fabian Johannes EBER
Aniela WOCHNER
Tilmann Roos
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Curevac SE
Original Assignee
Curevac SE
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Curevac SE filed Critical Curevac SE
Assigned to CUREVAC AG reassignment CUREVAC AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ROOS, TILMANN, EBER, Fabian Johannes, WOCHNER, Aniela
Publication of US20190100784A1 publication Critical patent/US20190100784A1/en
Assigned to CureVac SE reassignment CureVac SE CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: CUREVAC AG
Application granted granted Critical
Publication of US11920174B2 publication Critical patent/US11920174B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P19/00Preparation of compounds containing saccharide radicals
    • C12P19/26Preparation of nitrogen-containing carbohydrates
    • C12P19/28N-glycosides
    • C12P19/30Nucleotides
    • C12P19/34Polynucleotides, e.g. nucleic acids, oligoribonucleotides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6806Preparing nucleic acids for analysis, e.g. for polymerase chain reaction [PCR] assay
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6809Methods for determination or identification of nucleic acids involving differential detection
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2521/00Reaction characterised by the enzymatic activity
    • C12Q2521/30Phosphoric diester hydrolysing, i.e. nuclease
    • C12Q2521/327RNAse, e.g. RNAseH
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2525/00Reactions involving modified oligonucleotides, nucleic acids, or nucleotides
    • C12Q2525/10Modifications characterised by
    • C12Q2525/117Modifications characterised by incorporating modified base
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2525/00Reactions involving modified oligonucleotides, nucleic acids, or nucleotides
    • C12Q2525/10Modifications characterised by
    • C12Q2525/173Modifications characterised by incorporating a polynucleotide run, e.g. polyAs, polyTs
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2525/00Reactions involving modified oligonucleotides, nucleic acids, or nucleotides
    • C12Q2525/10Modifications characterised by
    • C12Q2525/204Modifications characterised by specific length of the oligonucleotides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2527/00Reactions demanding special reaction conditions
    • C12Q2527/125Specific component of sample, medium or buffer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2565/00Nucleic acid analysis characterised by mode or means of detection
    • C12Q2565/10Detection mode being characterised by the assay principle
    • C12Q2565/137Chromatographic separation
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B30/00ICT specially adapted for sequence analysis involving nucleotides or amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/03Phosphoric monoester hydrolases (3.1.3)
    • C12Y301/03001Alkaline phosphatase (3.1.3.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/04Phosphoric diester hydrolases (3.1.4)
    • C12Y301/04001Phosphodiesterase I (3.1.4.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/26Endoribonucleases producing 5'-phosphomonoesters (3.1.26)
    • C12Y301/26005Ribonuclease P (3.1.26.5)

Definitions

  • the present invention relates to the field of RNA analysis.
  • the invention provides a method suitable for use in quality control during or following production of RNA.
  • the method is particularly useful for the determination of critical RNA quality attributes including RNA identity, RNA integrity, capping efficiency, poly(A) tail length, RNA oxidation, and the incorporation efficiency and amount of modified nucleotides.
  • RNA molecules represent an emerging class of drugs. RNA-based therapeutics may be used in immunotherapy, gene therapy and genetic vaccination, belonging to the most promising and quickly developing therapeutic fields of modern medicine. RNA-based therapeutics may provide highly specific and individual treatment options for therapy of a large variety of diseases.
  • RNA-based therapeutics include mRNA molecules encoding antigens for use as vaccines (Fotin-Mleczek et al. (2012) J. Gene Med. 14(6):428-439).
  • RNA molecules for replacement therapies, e.g. providing missing proteins such as growth factors or enzymes to patients.
  • non-coding immunostimulatory RNA molecules and other non-coding RNAs such as microRNAs, siRNAs, CRISPR/Cas9 guide RNAs, and long non-coding RNAs is considered.
  • Short RNA molecules can be synthesized by chemical methods, whereas long RNAs are typically produced by in vitro transcription using suitable DNA templates with a promoter and RNA polymerases, for example bacteriophage SP6, T3 or T7 RNA polymerases which are able to bind to said promoter.
  • RNA polymerases for example bacteriophage SP6, T3 or T7 RNA polymerases which are able to bind to said promoter.
  • RNA molecules for the successful development of RNA-based therapeutics, the production of RNA molecules as active pharmaceutical ingredients must be efficient in terms of yield, quality, safety and costs, especially when RNA is produced at a large scale.
  • quality controls may be implemented during RNA production and/or following RNA production and/or as a batch release quality control.
  • RNA For any application of RNA in a scientific or therapeutic setting, it is highly desired or mandatory to use RNA with a defined sequence that can be reproduced in a reliable manner.
  • the 5′ terminal as well as the 3′ terminal region of a protein coding RNA molecule are known to be involved in the regulation of the mRNA stability and translation efficiency.
  • the 5′ cap structure and the 3′ poly(A) tail are important features for the efficient translation of mRNA and protein synthesis in eukaryotic cells and, therefore, the produced RNA should be controlled for such key functional features.
  • RNA molecules it is desired to control the composition of the drug. Therefore, it is highly desired or mandatory to control the identity and/or integrity of the RNA molecules. Moreover, it is desired to control the produced RNA (in case of protein coding mRNA) for capping efficiency, and for the quality and length of the poly(A) tail or other homopolymeric sequence elements (e.g., poly(C) sequence). Furthermore, it could be shown that oxidation of RNA can lead to translational errors (Kumaya et al. (2006) Nucleic Acid Symposium Series 50: 99-100). Therefore it is desired to have a quality control to determine the oxidation status of the produced RNA. Additionally it is desirable to determine the incorporation efficiency and amount of modified nucleotides if used for RNA in vitro transcription.
  • RNA identity can be analyzed using PCR based methods or sequencing.
  • RNA integrity can be analyzed by electrophoretic methods.
  • quality controls have been established in the art to analyze the capping efficiency (WO 2015/01416; Theus et al. (1990) Biotechniques Rapid Dispatches 9(5): 610-612) or to analyze poly(A) tails at the 3′ end of an RNA (PCT/EP 2015/001336).
  • HPLC-based methods have been described for determining the degree of RNA oxidation and the incorporation of modified nucleotides of cellular RNAs (Nees et al. (2014) Innate DNA and RNA Recognition, methods in Molecular Biology, vol. 1169: 3-14).
  • the percentage of modified nucleosides can also be determined by separating the nucleosides by HPLC and subsequent quantification using GC/MS or LC/MS (Tanaka et al. (2012) Free Radical Biology and Medicine 53(4): 854-862; Su et al. (2014) Nature Protocols 9(4): 828-841).
  • RNA identity RNA identity
  • RNA integrity RNA integrity
  • RNA capping efficiency RNA polyadenylation
  • RNA oxidation RNA oxidation
  • RNA species comprising a poly(A) tail and a cap-structure, produced via RNA in vitro transcription, was hydrolyzed to individual nucleoside monophosphates. After dephosphorylation, the nucleosides A, C, m7G (cap analogue), G, U and 8-OH-GTP (oxidized GTP) were separated and quantified by HPLC.
  • the inventors determined the identity and the molarity of the RNA (comprising a poly(A) tail). In addition, the quantification of the m7G cap analogue allowed for a calculation of the capping degree of the RNA.
  • the length of the poly(A) tail was determined.
  • the quality of other homopolymeric elements such as a poly(C) sequence can be determined.
  • the amount of modified nucleotides the frequency of incorporation and the amount of modified nucleotides was calculated.
  • the oxidation status of the RNA was determined by detecting and quantifying oxidized nucleosides.
  • the present invention relates to a method for analyzing a sample comprising RNA molecules transcribed from a template DNA with known sequence and length or chemically synthesized with known sequence and length, comprising the steps of:
  • Step d) may further comprise determining at least one of:
  • Step d) may also comprise determining:
  • RNA molecules may be provided by chemical synthesis or by in vitro transcription.
  • the method may further comprise a step al) of purifying the RNA molecules, preferably by HPLC.
  • RNA molecules may be completely hydrolyzed by enzyme treatment, preferably by treatment with a nuclease, a phosphatase and a phosphodiesterase.
  • the nuclease is Nuclease P1 from Penicillium citrinum
  • the phosphodiesterase is Phosphodiesterase I from Crotalus adamanteus and/or the phosphatase is shrimp alkaline phosphatase.
  • step c) the nucleosides may be separated using an octadecyl capped silica column.
  • a mobile phase comprising an aqueous solvent is used in the HPLC of step c).
  • the aqueous solvent is ammonium acetate and more preferably, the aqueous solvent is 5 mM NH 4 OAc, pH 6.
  • the nucleosides may be eluted with an organic solvent, preferably with a linear gradient of said organic solvent. More preferably, the gradient is from 0% to 20% of the organic solvent.
  • the organic solvent is 40% v/v acetonitrile.
  • Step c) may further comprise detecting the released nucleosides and identifying the released nucleosides by comparison to a standard for each nucleoside.
  • the released nucleosides are quantified by determining the peak area for each of the nucleosides and comparing it to a standard curve.
  • Step d) may comprise determining the matching of the sequence of the RNA molecules with the known sequence and/or the matching of the length of the RNA molecules with the known length by comparing the measured ratio of a specific nucleoside in the sample with the expected ratio of said nucleoside.
  • the measured and the expected ratio may be determined for each of C, G, U and/or A.
  • a mismatch between the values for rm and re indicates no matching of the sequence of the RNA molecules with the known sequence and/or no matching of the length of the RNA molecules with the known length.
  • the cap analogue may be selected from the group consisting of: G[5]ppp[5]G, m7G[5′]ppp[5]G, m32,2,7G[5]ppp[5]G, m27,3′-OG[5]ppp[5]G (3′-ARCA), m27,2′-OGpppG (2′-ARCA), m27,2′-OGppspG D1 ( ⁇ -S-ARCA D1) and m27,2′-OGppspG D2 ( ⁇ -S-ARCA D2).
  • the number of copies of the RNA molecules in the sample may be determined as described above for polyadenylated and not polyadenylated RNA molecules.
  • the number of copies of the RNA molecules in the sample may be determined as described above.
  • the modified nucleoside may be selected from the group consisting of: 2-amino-6-chloropurineriboside-5′-triphosphate, 2-Aminopurine-riboside-5′-triphosphate; 2-aminoadenosine-5′-triphosphate, 2′-Amino-2′-deoxycytidine-triphosphate, 2-thiocytidine-5′-triphosphate, 2-thiouridine-5′-triphosphate, 2′-Fluorothymidine-5′-triphosphate, 2′-O-Methyl inosine-5′-triphosphate 4-thiouridine-5′-triphosphate, 5-aminoallylcytidine-5′-triphosphate, 5-aminoallyluridine-5′-triphosphate, 5-bromocytidine-5′-triphosphate, 5-bromouridine-5′-triphosphate, 5-Bromo-2′-deoxycytidine-5′-triphosphate, 5-Bromo-2′-deoxycy
  • the modified nucleosides may be oxidized nucleosides and % mN may indicate the oxidation status of the RNA molecules.
  • the oxidized nucleosides may be selected from the group consisting of: 8-hydroxyguanosine (8-OHG), 8-oxo-7,8-dihydro-2′-deoxyguanosine, dehydroguanidinohyantoin, 2,4,6-trioxo-[1,3,5]triazinane-1-carboxamidine, N-nitro-dehydroguanidinohydantoin, spiroiminodihydantoin, guanidinohydantoin, 4-hydroxy-2,5-dioxo-imidazolidine-4-carboxylic acid, 3-nitrotyrosine, 3.3′-dityrosine, 3-hydroxy-5-imino-3,3a,4,5-tetrahydro-1H-imidazo[4,5-d]imidazol-2-one, parabanic acid, cyanuric acid, 4-hydroxy-8-oxo-4,8-dihydro-2′-desoxyguanosine, 4,5-
  • the RNA molecules are mRNA molecules.
  • the method may additionally comprise step al) of modifying the RNA molecules.
  • the modifying of the RNA molecules may comprise enzymatically adding a poly(A) stretch to the RNA molecules.
  • the number of copies of the RNA molecules in the sample may be determined as described above.
  • the modifying of the RNA molecules may also comprise enzymatically adding a cap structure to the RNA molecules.
  • the number of copies of the RNA molecules in the sample may be determined as described above.
  • the modifying of the RNA molecules may also comprise enzymatically modifying the nucleotides of the RNA molecules.
  • the present invention also relates to the use of the method according to the invention in the quality control of RNA prepared by RNA in vitro transcription or by chemical synthesis.
  • FIG. 1 shows an exemplary chromatogram of the hydrolysate of PpLuc mRNA (R2988) and respective chromatograms of standards for calibration.
  • the following chromatograms are shown (from top to bottom): PpLuc hydrolysate (A), GTP standard (B), UTP standard (C), CTP standard (D), ATP standard (E), 8-OH-GTP standard (F) and m7GpppG cap analogue standard (G).
  • the arrows show how corresponding peaks in the hydrolysate were identified using the standards. For experimental details see Example 3.
  • FIG. 2 shows a comparison of the measured and the expected nucleoside ratios (C:G:U) for the indicated five mRNA hydrolysates.
  • C:G:U expected nucleoside ratios
  • FIG. 3 shows a comparison of the capping degree calculated for the indicated three mRNA hydrolysates. For experimental details see Example 3.
  • FIG. 4 shows a comparison of the length of the poly(A)stretch in the indicated five mRNA hydrolysates. For experimental details see Example 3.
  • FIG. 5 shows a comparison of the frequency of 8-OH-G incorporation in PpLuc mRNA without and with 8-OH-GTP added to the respective transcription reaction [“PpLuc” and “PpLuc (8-OH-G)” respectively].
  • RNA is the usual abbreviation for ribonucleic acid. It is a nucleic acid molecule, i.e. a polymer consisting of nucleotide monomers. These nucleotides are usually adenosine monophosphate (AMP), uridine monophosphate (UMP), guanosine monophosphate (GMP) and cytidine monophosphate (CMP) monomers or analogues thereof, which are connected to each other along a so-called backbone.
  • the backbone is formed by phosphodiester bonds between the sugar, i.e. ribose, of a first and a phosphate moiety of a second, adjacent monomer.
  • RNA sequence The specific order of the monomers, i.e. the order of the bases linked to the sugar/phosphate-backbone, is called the RNA sequence.
  • RNA may be obtainable by transcription of a DNA sequence, e.g., inside a cell. In eukaryotic cells, transcription is typically performed inside the nucleus or the mitochondria. In vivo, transcription of DNA usually results in the so-called premature RNA (also called pre-mRNA, precursor mRNA or heterogeneous nuclear RNA) which has to be processed into so-called messenger RNA, usually abbreviated as mRNA. Processing of the premature RNA, e.g.
  • RNA in eukaryotic organisms, comprises a variety of different posttranscriptional modifications such as splicing, 5′-capping, polyadenylation, export from the nucleus or the mitochondria and the like.
  • the sum of these processes is also called maturation of RNA.
  • the mature messenger RNA usually provides the nucleotide sequence that may be translated into an amino acid sequence of a particular peptide or protein.
  • a mature mRNA comprises a 5′-cap, optionally a 5′UTR, an open reading frame, optionally a 3′UTR and a poly(A) tail.
  • RNA In addition to messenger RNA, several non-coding types of RNA exist which may be involved in regulation of transcription and/or translation, and immunostimulation.
  • RNA further encompasses any type of single stranded (ssRNA) or double stranded RNA (dsRNA) molecule known in the art, such as viral RNA, retroviral RNA and replicon RNA, small interfering RNA (siRNA), antisense RNA (asRNA), circular RNA (circRNA), CRISPR/Cas9 guide RNA, ribozymes, aptamers, riboswitches, immunostimulating RNA, transfer RNA (tRNA), ribosomal RNA (rRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), microRNA (miRNA), and Piwi-interacting RNA (piRNA).
  • ssRNA single stranded
  • dsRNA double stranded RNA
  • viral RNA small interfering
  • sample comprising RNA molecules is intended to include a sample comprising a population of RNA molecules which have been transcribed from the same template DNA of known sequence.
  • a sample comprising RNA molecules will therefore comprise many RNA molecules of the same sequence, although any modifications including polyadenylation, capping, incorporation of modified nucleotides may not occur at the same rate on all of them so that the final structure of the RNA molecules within the sample may not be the same for all the RNA molecules.
  • the sample comprising RNA molecules does not comprise RNA molecules which have been transcribed from two or more different DNA templates.
  • Expected sequence refers to the sequence identical to the known sequence of the template used for RNA in vitro transcription or the sequence used for chemical synthesis, i.e. the desired sequence.
  • Expected length refers to the length of the RNA molecule which could be expected from the known length of the template used for RNA in vitro transcription or the sequence used for chemical synthesis, i.e. the desired length.
  • RNA hydrolysis is a reaction in which a phosphodiester bond in the sugar-phosphate backbone of RNA is broken, thereby cleaving the RNA molecule into shorter stretches.
  • RNA is susceptible to hydrolysis because the ribose sugar in RNA has a hydroxyl group at the 2′ position. This feature makes RNA chemically unstable compared to DNA, which does not have this 2′ OH group and thus is not susceptible to base-catalyzed hydrolysis.
  • RNA hydrolysis occurs when the deprotonated 2′ OH of the ribose, acting as a nucleophile, attacks the adjacent phosphorus in the phosphodiester bond of the sugar-phosphate backbone of the RNA, resulting in a transition state in which the phosphorus is bonded to five oxygen atoms. The phosphorus then detaches from the oxygen connecting it to the adjacent sugar, resulting in ester cleavage of the RNA backbone. This produces a 2′,3′-cyclic phosphate that can then yield either a 2′- or a 3′-nucleotide when hydrolyzed. This hydrolysis can occur spontaneously, then called auto-hydrolysis. RNA hydrolysis is increased if the pH of the RNA solution is alkaline (alkaline hydrolysis).
  • the RNA may be hydrolyzed enzymatically by the action of phosphodiesterases, which are capable of breaking the phosphodiester bonds that make up the backbone of the RNA molecule.
  • Complete hydrolysis is achieved when the RNA molecule has been hydrolyzed to an extent that no phosphodiester bridges remain between nucleotides, i.e. the backbone of the RNA molecule has been completely disassembled into single nucleotides. Hence, no longer nucleotide stretches comprising two or more nucleotides linked by phosphodiester bridges can be detected.
  • Hydrolysis of an RNA molecule can be achieved by enzymatic hydrolysis (e.g., using nucleases, phosphodiesterases) or e.g.
  • alkaline hydrolysis for example by incubating an RNA sample in alkaline hydrolysis buffer (e.g., 50 mM sodium carbonate [NaHCO 3 /Na 2 CO 3 ] pH 9.2, 1 mM EDTA) at 95° C. until complete hydrolysis has occurred.
  • alkaline hydrolysis buffer e.g., 50 mM sodium carbonate [NaHCO 3 /Na 2 CO 3 ] pH 9.2, 1 mM EDTA
  • complete hydrolysis also includes treatment with a phosphatase that dephosphorylates the released, single nucleotides into single nucleosides.
  • Phosphodiesterase A phosphodiesterase (PDE) is any enzyme that breaks a phosphodiester bond.
  • Phosphodiesterase families include cyclic nucleotide phosphodiesterases, phospholipases C and D, autotaxin, sphingomyelin phosphodiesterase, DNases, RNases, and restriction endonucleases, as well as numerous less-well-characterized small-molecule phosphodiesterases. Of these, several can catalyze cleavage at specific sites on an RNA molecule by breaking the phosphodiester backbone of the molecule, for example nucleases and ribonucleolytic ribozymes.
  • Suitable phosphodiesterases for use in the present invention include phosphodiesterase I from Crotalus adamanteus, phosphodiesterase I from Bothrops atrox , phosphodiesterase I from Crotalus atrox and phosphodiesterase from bovine spleen.
  • a preferred phosphodiesterase for use in the present invention is phosphodiesterase I from Crotalus adamanteus (available from Sigma Aldrich).
  • Nuclease A nuclease is a phosphodiesterase capable of cleaving the phosphodiester bonds between the nucleotide subunits of nucleic acids. Nucleases are usually further divided into endonucleases, which cleave phosphodiester bonds within a nucleotide chain, and exonucleases, which cleave phosphodiester bonds at either terminus of the nucleic acid, although some of the enzymes may fall in both categories. Well known nucleases are deoxyribonuclease and ribonuclease.
  • Suitable nucleases for use in the present invention include benzonase, nuclease P1 from Penicillium citrinum , nuclease S1 from Aspergillus oryzae , ribonuclease A from bovine pancreas and ribonuclease T1 from Aspergillus oryzae .
  • a preferred nuclease for use in the present invention is nuclease P1 from Penicillium citrinum (available from Sigma Aldrich).
  • Exoribonucleases degrade RNA from the 5′ or 3′ end of the RNA molecule. These enzymes are involved in the degradation of many different RNA species, including mRNA, tRNA, rRNA and miRNA. Exoribonucleases can be single proteins (such as Nuclease P1 from Penicillium citrinum and PDE1 from Crotalus adamanteus, 5′ ⁇ 3′ and 3′ ⁇ 5′ exoribonucleases, respectively) or complexes of multiple proteins, such as the exosome complex, in which four of the major exoribonuclease families are represented.
  • Single proteins such as Nuclease P1 from Penicillium citrinum and PDE1 from Crotalus adamanteus, 5′ ⁇ 3′ and 3′ ⁇ 5′ exoribonucleases, respectively
  • complexes of multiple proteins such as the exosome complex, in which four of the major exoribonuclease families are represented.
  • a phosphatase is an enzyme that removes a phosphate group from its substrate by hydrolyzing phosphoric acid monoesters into a phosphate ion and a molecule with a free hydroxyl group. Examples include alkaline phosphatase, deoxyribonucleotide phosphatases, ribonucleotide phosphatases, pyrophosphatases, and protein phosphatases, which remove a phosphate group from the phosphorylated amino acid residue of the substrate protein.
  • the phosphatase is preferably an alkaline phosphatase, such as shrimp alkaline phosphatase, calf-intestinal alkaline phosphatase, placental alkaline phosphatase, and its C terminally truncated version secreted alkaline phosphatase.
  • alkaline phosphatase such as shrimp alkaline phosphatase, calf-intestinal alkaline phosphatase, placental alkaline phosphatase, and its C terminally truncated version secreted alkaline phosphatase.
  • shrimp alkaline phosphatase available from New England Biolabs.
  • HPLC is the common abbreviation of the term “high performance liquid chromatography”.
  • HPLC high performance liquid chromatography
  • RP-HPLC reversed phase HPLC
  • size exclusion chromatography gel filtration, affinity chromatography, hydrophobic interaction chromatography or ion pair chromatography, wherein reversed phase HPLC is preferred.
  • Reversed phase HPLC uses a non-polar stationary phase and a moderately polar mobile phase and therefore is based on hydrophobic interactions which result from repulsive forces between a relatively polar solvent, the relatively non-polar analyte, and the non-polar stationary phase (reversed phase principle).
  • the retention time on the column is therefore longer for molecules which are more non-polar in nature, allowing polar molecules to elute more readily.
  • the retention time is increased by the addition of polar solvent to the mobile phase and decreased by the addition of more hydrophobic solvent.
  • RNA molecule as an analyte may play an important role in its retention characteristics.
  • an analyte having more apolar functional groups results in a longer retention time because it increases the molecule's hydrophobicity and therefore the interaction with the non-polar stationary phase.
  • Very large molecules can result in incomplete interaction between the large analyte surface and the alkyl chain. Retention time increases with hydrophobic surface area which is roughly inversely proportional to solute size. Branched chain compounds elute more rapidly than their corresponding isomers because the overall surface area is decreased.
  • Separation/separating refers to the separation of all different nucleosides U, C, G and possibly A present in a nucleic acid molecule from each other based on their differential interactions with the adsorbent material due to their various characteristics such as, for example, size, charge, branching, or apolar or polar functional groups.
  • the more a nucleoside interacts with the adsorbent material the longer it is retained, i.e. the longer is its retention time. Based on their retention time, nucleosides will therefore elute at different times, and can be segregated into separate, distinct fractions that will only contain one type of nucleoside. Thereby, each of the separated nucleosides can be identified and quantified accurately.
  • RNA molecules in a sample is the number of copies of said molecules present in the sample.
  • RNA molecules During quality control of RNA production it is crucial to demonstrate that the sequence of the produced RNA molecules corresponds to or matches with the desired or expected or known sequence of the RNA molecule, i.e. that the RNA in vitro transcription or chemical synthesis of the RNA worked and did not produce any errors. This sequence identity of the RNA molecules can either be assessed according to the method disclosed herein or by e.g. sequencing.
  • Integrity of RNA molecules The term “integrity” describes whether the complete RNA molecule is obtained and maintained. Accordingly, the integrity can be determined by measuring the length of the RNA molecule. If the complete RNA molecule is obtained, the RNA molecule obtained will have the expected or known length. If an incomplete RNA molecule is obtained, the length of the RNA molecule will be different from the expected or known length. Low integrity or differences in the length of the RNA molecule could be due to, amongst others, degradation, cleavage, incorrect basepairing, lack of or incomplete capping, lack of or incomplete polyadenylation, or incomplete transcription. This will become evident in a mismatch between measured and known, expected nucleoside ratios for the RNA molecules.
  • RNA molecules having a different length as the expected length but the same sequence as the expected or known sequence
  • RNA molecules having the same length as the expected or known length but a sequence different from the expected sequence.
  • a 5′ cap is typically a modified nucleotide, particularly a guanine nucleotide, added to the 5′ end of an RNA molecule.
  • the 5′ cap is added using a 5′-5′-triphosphate linkage.
  • a 5′ cap may be methylated, e.g. m7GpppN, wherein N is the terminal 5′ nucleotide of the nucleic acid carrying the 5′ cap, typically the 5′-end of an RNA.
  • the naturally occurring 5′ cap is m7GpppN (cap 0).
  • 5′cap structures include glyceryl, inverted deoxy abasic residue (moiety), 4′,5′ methylene nucleotide, 1-(beta-D-erythrofuranosyl) nucleotide, 4′-thio nucleotide, carbocyclic nucleotide, 1,5-anhydrohexitol nucleotide, L-nucleotides, alpha-nucleotide, modified base nucleotide, threo-pentofuranosyl nucleotide, acyclic 3′,4′-seco nucleotide, acyclic 3,4-dihydroxybutyl nucleotide, acyclic 3,5 dihydroxypentyl nucleotide, 3′-3′-inverted nucleotide moiety, 3′-3′-inverted abasic moiety, 3′-2′-inverted nucleotide moiety, 3′-2′-inverted abasic
  • 5′ cap structures can be further methylated, e.g. cap 1 (methylation of the ribose of the adjacent nucleotide of m7G), cap 2 (methylation of the ribose of the 1 st and 2 nd nucleotide downstream of the m7G), cap 3 (methylation of the ribose of the 1 st and 2 nd and 3 rd nucleotide downstream of the m7G) and cap 4 (methylation of the ribose of the 1 st , 2 nd , 3 rd and 4 th nucleotide downstream of the m7G).
  • cap 1 methylation of the ribose of the adjacent nucleotide of m7G
  • cap 2 methylation of the ribose of the 1 st and 2 nd nucleotide downstream of the m7G
  • cap 3 methylation of the ribose of the 1 st and 2 nd and 3 rd
  • a 5′ cap structure may be formed by a cap analogue.
  • a 5′ cap structure may be formed in chemical RNA synthesis or RNA in vitro transcription (co-transcriptional capping) or a cap structure may be formed in vitro using capping enzymes (e.g., commercially available capping kits).
  • a cap analogue refers to a non-polymerizable di-nucleotide that has cap functionality in that it facilitates translation or localization, and/or prevents degradation of the RNA molecule when incorporated at the 5′ end of the RNA molecule.
  • Non-polymerizable means that the cap analogue will be incorporated only at the 5′terminus because it does not have a 5′ triphosphate and therefore cannot be extended in the 3′ direction by a template-dependent RNA polymerase.
  • Cap analogues include, but are not limited to, a chemical structure selected from the group consisting of m7GpppG, m7GpppA, m7GpppC; unmethylated cap analogues (e.g., GpppG); dimethylated cap analogue (e.g., m2,7GpppG), trimethylated cap analogue (e.g., m2,2,7GpppG), dimethylated symmetrical cap analogues (e.g., m7Gpppm7G), or anti reverse cap analogues (e.g., ARCA; m7,2′OmeGpppG, m7,2′dGpppG, m7,3′OmeGpppG, m7,3′dGpppG and their tetraphosphate derivatives) (Stepinski et al., 2001. RNA 7(10):1486-95). Examples of cap analogues are shown in Table 1.
  • Triphosphate cap analogue Tetraphosphate cap analogue m 7 Gp 3 G m 7 Gp 4 G m 2 7,3′-O Gp 3 G b 7 Gp 4 G b 7 Gp 3 G b 7 m 3′-O Gp 4 G e 7 Gp 3 G m 2 2,7 Gp 4 G m 2 2,7 Gp 3 G m 3 2,2,7 Gp 4 G m 3 2,2,7 Gp 3 G b 7 m 2 Gp 4 G m 7 Gp 3 2′dG m7Gp 4 m 7 G m 7 Gp 3 m 2′-O G m 7 Gp 3 m 7 G m 2 7,2′-O Gp 3 G m 2 7,2′-O Gp 3 GpppsG (D1) m 2 7,2′-O GpppsG (D2) m 2 7,2′-O GppspG (D1) m 2 7,2′-O GppspG (D2) m 2 7,2′-O G
  • cap analogues are G[5′]ppp[5′]G, m 7 G[5′]ppp[5′]G, m3 2,2,7 G[5′]ppp[5′]G, m2 7,3′-O G[5′]ppp[5′]G (3′-ARCA), m2 7,2′-O GpppG (2′-ARCA), m2 7,2′-O GppspG D1 ( ⁇ -S-ARCA D1) and m 2 7,2′-O GppspG D2 ( ⁇ -S-ARCA D2).
  • Capping degree The relative amount (in percent) of capped RNA molecules with respect to the total amount of RNA molecules in the sample is also referred to herein as “capping degree”.
  • the relative amount (in percent) of capped and non-capped RNA molecules to be analyzed is calculated using the following formulas 4.1 and 4.2:
  • Homopolymeric elements are stretches (sequences) in an RNA molecule consisting entirely or almost entirely of a single, repeated nucleotide.
  • the most prominent homopolymeric element in RNA is the poly(A) tail at the 3′ end of eukaryotic mRNA.
  • polyadenylation, or addition of a poly(A) tail to an mRNA is part of the process that produces mature mRNA for translation. It therefore forms part of the process of gene expression. The process of polyadenylation begins as the transcription of a gene terminates.
  • the 3′-most segment of the newly made pre-mRNA is first cleaved off by a set of proteins; these proteins then synthesize the poly(A) tail at the RNA's 3′ end.
  • these proteins may add a poly(A) tail at any one of several possible sites. Therefore, polyadenylation can produce more than one molecule from a single transcript (alternative polyadenylation), similar to alternative splicing.
  • the poly(A) tail is important for the nuclear export, translation, and stability of mRNA.
  • the tail is shortened over time, and, when it is short enough, the mRNA is enzymatically degraded.
  • mRNAs with short poly(A) tails are stored for later activation by re-polyadenylation in the cytosol.
  • polyadenylation occurs in bacteria, it promotes RNA degradation. This is also sometimes the case for eukaryotic non-coding RNAs.
  • mRNA molecules in both prokaryotes and eukaryotes have polyadenylated 3′-ends, with the prokaryotic poly(A) tails generally being shorter and present on fewer mRNA molecules.
  • poly(A) polymerase which uses ATP as a substrate for template-independent addition of adenosine monophosphates to the 3′-hydroxyl terminus of RNA molecules. They typically produce a poly(A)-tail length of ⁇ 150 bases, but can be adapted to adjust the length of the poly(A)-tails generated.
  • poly(A) tails can be produced during RNA in vitro transcription, if the DNA template used for in vitro transcription encodes a poly(A) tail or a poly(A) tail can be generated by chemical synthesis.
  • a poly(C) stretch, or C-rich stability element is understood to be a sequence of cytosine residues, e.g., of up to about 400 nucleotides, e.g. from about 20 to about 400, preferably from about 50 to about 400, more preferably from about 50 to about 300, even more preferably from about 50 to about 250, most preferably from about 60 to about 250 nucleotides, which is preferably added to the 3′-terminus of an RNA, in particular mRNA.
  • a poly(C) sequence is typically located at the 3′-end of an (m)RNA, but 5′ of any poly(A) tail.
  • Eukaryotic mRNAs can have half-lives that vary from as short as several minutes to as long as several days. In differentiating human erythroid cells the deposition of high levels of alpha globin protein is dependent on an unusually long half-life of the alpha globin mRNA (greater than 24 hrs).
  • the stability determinant for alpha globin mRNA has been mapped to a pyrimidine-rich segment of its 3′ UTR. This poly(C) stretch confers stability by directing the assembly of a specific alpha ribonucleoprotein complex at this site.
  • homopolymeric elements include poly(U) stretches that are known from viral RNA (e.g., coronavirus).
  • Modified nucleoside refers to chemical modifications comprising backbone modifications as well as sugar modifications or base modifications. These modified nucleosides are herein also called (nucleotide) analogues.
  • the modified nucleosides as defined herein are nucleotide analogues/modifications, e.g. backbone modifications, sugar modifications or base modifications.
  • a backbone modification in connection with the present invention is a modification in which phosphates of the backbone of the nucleotides are chemically modified.
  • a sugar modification in connection with the present invention is a chemical modification of the sugar of the nucleotides.
  • a base modification in connection with the present invention is a chemical modification of the base moiety of the nucleotides.
  • nucleotide analogues or modifications are preferably selected from nucleotide analogues which are applicable for transcription and/or translation.
  • modified nucleosides and nucleotides which may be used in the context of the present invention can be modified in the sugar moiety.
  • the 2′ hydroxyl group (OH) can be modified or replaced with a number of different “oxy” or “deoxy” substituents.
  • R alkoxy or aryloxy
  • R alkoxy or ary
  • “Deoxy” modifications include hydrogen, amino (e.g. NH 2 ; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid); or the amino group can be attached to the sugar through a linker, wherein the linker comprises one or more of the atoms C, N, and O.
  • the sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose.
  • a modified nucleotide can include nucleotides containing, for instance, arabinose as the sugar.
  • the phosphate backbone may further be modified in the modified nucleosides and nucleotides.
  • the phosphate groups of the backbone can be modified by replacing one or more of the oxygen atoms with a different substituent.
  • the modified nucleosides and nucleotides can include the full replacement of an unmodified phosphate moiety with a modified phosphate as described herein. Examples of modified phosphate groups include, but are not limited to, phosphorothioate, phosphoroselenates, borano phosphates, borano phosphate esters, hydrogen phosphonates, phosphoroamidates, alkyl or aryl phosphonates and phosphotriesters.
  • Phosphorodithioates have both non-linking oxygens replaced by sulfur.
  • the phosphate linker can also be modified by the replacement of a linking oxygen with nitrogen (bridged phosphoroamidates), sulfur (bridged phosphorothioates) and carbon (bridged methylene-phosphonates).
  • modified nucleosides and nucleotides which may be used in the present invention, can further be modified in the nucleobase moiety.
  • nucleobases found in RNA include, but are not limited to, adenine, guanine, cytosine and uracil.
  • nucleosides and nucleotides described herein can be chemically modified on the major groove face.
  • the major groove chemical modifications can include an amino group, a thiol group, an alkyl group, or a halo group.
  • the nucleotide analogues/modifications are selected from base modifications, which are preferably selected from 2-amino-6-chloropurineriboside-5′-triphosphate, 2-Aminopurine-riboside-5′-triphosphate; 2-aminoadenosine-5′-triphosphate, 2′-Amino-2′-deoxycytidine-triphosphate, 2-thiocytidine-5′-triphosphate, 2-thiouridine-5′-triphosphate, 2′-Fluorothymidine-5′-triphosphate, 2′-O-Methyl inosine-5′-triphosphate 4-thiouridine-5′-triphosphate, 5-aminoallylcytidine-5′-triphosphate, 5-amino allyluridine-5′-triphosphate, 5-bromocytidine-5′-triphosphate, 5-bromouridine-5′-triphosphate, 5-Bromo-2′-deoxycytidine-5′-
  • nucleotides for base modifications selected from the group of base-modified nucleotides consisting of 5-methylcytidine-5′-triphosphate, 7-deazaguanosine-5′-triphosphate, 5-bromocytidine-5′-triphosphate, and pseudouridine-5′-triphosphate.
  • modified nucleosides include pyridin-4-one ribonucleoside, 5-aza-uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyluridine, 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uridine, 1-taurinomethyl-4-thio-uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-1-methyl-pseudouridine, 2-thio-1-methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-methyl-1-
  • modified nucleosides include 5-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine, N4-acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5-hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-1-methyl-pseudoisocytidine, 4-thio-1-methyl-1-deaza-pseudoisocytidine, 1-methyl-1-deaza-pseudo iso cytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine, 2-thio-ze
  • modified nucleosides include 2-aminopurine, 2,6-diaminopurine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, 2-methylthio-N6-(cis-hydroxyisopentenyl) adenosine, N6-glycinylcarbamoyladenosine, N6-threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6-di
  • modified nucleosides include inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7-methylinosine, 6-methoxy-guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2-dimethylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, l-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine.
  • the nucleoside can be modified on the major groove face and can include replacing hydrogen on C-5 of uracil with a methyl group or a halo group.
  • a modified nucleoside is 5′-O-(1-Thiophosphate)-Adenosine, 5′-O-(1-Thiophosphate)-Cytidine, 5′-O-(1-Thiophosphate)-Guano sine, 5′-O-(1-Thiophosphate)-Uridine or 5′-O-(1-Thiophosphate)-Pseudouridine.
  • the modified nucleosides include nucleoside modifications selected from 6-aza-cytidine, 2-thio-cytidine, ⁇ -thio-cytidine, Pseudo-iso-cytidine, 5-aminoallyl-uridine, 5-iodo-uridine, N1-methyl-pseudouridine, 5,6-dihydrouridine, ⁇ -thio-uridine, 4-thio-uridine, 6-aza-uridine, 5-hydroxy-uridine, deoxy-thymidine, 5-methyl-uridine, Pyrrolo-cytidine, inosine, ⁇ -thio-guanosine, 6-methyl-guanosine, 5-methyl-cytdine, 8-oxo-guanosine, 7-deaza-guanosine, N1-methyl-adenosine, 2-amino-6-Chloro-purine, N6-methyl-2-amino-purine, Pseudo-is
  • RNA modifications may occur by oxidation of the RNA molecule through reactive oxygen species which are involved in the killing of bacteria and in cell signaling pathways. More than 20 different purine and pyrimidine modifications formed by reactive oxygen species are known, which include: 8-hydroxyguanosine (8-OHG), which can react further to 8-oxo-7,8-dihydro-2′-deoxyguanosine, dehydroguanidinohyantoin, 2,4,6-trioxo-[1,3,5]triazinane-1-carboxamidine, N-nitro-dehydroguanidinohydantoin, spiroiminodihydantoin, guanidinohydantoin, 4-hydroxy-2,5-dioxo-imidazolidine-4-carboxylic acid, 3-nitrotyrosine, 3.3′-dityrosine, 3-hydroxy-5-imino-3,3a,4,5-tetrahydro-1H-imid
  • RNA molecules Determining the percentage of modified nucleosides that arise from oxidation in an RNA molecule will therefore provide information on the oxidation status of the RNA molecules.
  • the most prominent oxidative modification is 8-hydroxyguanosine.
  • 8-OHG modifications in mRNA lead to reduced protein levels and altered protein function due to ribosome stalling (Shan et al. (2007) FASEB J. 21: 2753-2764).
  • age-associated oxidative damage to RNA has been demonstrated in neurons and may play a role in neurodegeneration and other diseases (Nunomura et al. (2012) Neurotox. Res. 22: 231-248).
  • Chemical synthesis of RNA Chemical synthesis of relatively short fragments of oligonucleotides with defined, known chemical structure provides a rapid and inexpensive access to custom-made oligonucleotides of any desired sequence. Whereas enzymes synthesize DNA and RNA only in the 5′ to 3′ direction, chemical oligonucleotide synthesis does not have this limitation, although it is most often carried out in the opposite, i.e. the 3′ to 5′ direction.
  • the process is implemented as solid-phase synthesis using the phosphoramidite method and phosphoramidite building blocks derived from protected nucleosides (A, C, G, and U), or chemically modified nucleosides.
  • the building blocks are sequentially coupled to the growing oligonucleotide chain on a solid phase in the order required by the sequence of the product in a fully automated process.
  • the product is released from the solid phase to the solution, deprotected, and collected.
  • the occurrence of side reactions sets practical limits for the length of synthetic oligonucleotides (up to about 200 nucleotide residues), because the number of errors increases with the length of the oligonucleotide being synthesized.
  • Products are often isolated by HPLC to obtain the desired oligonucleotides in high purity.
  • oligonucleotides find a variety of applications in molecular biology and medicine. They are most commonly used as antisense oligonucleotides, small interfering RNA, primers for DNA sequencing and amplification, probes for detecting complementary DNA or RNA via molecular hybridization, tools for the targeted introduction of mutations and restriction sites, and for the synthesis of artificial genes.
  • RNA in vitro transcription relate to a process wherein RNA is synthesized in a cell-free system (in vitro).
  • DNA particularly plasmid DNA
  • RNA is used as template of known sequence and length for the generation of RNA transcripts.
  • RNA may be obtained by DNA-dependent in vitro transcription of an appropriate DNA template, which according to the present invention is preferably a linearized plasmid DNA template.
  • the promoter for controlling in vitro transcription can be any promoter for any DNA-dependent RNA polymerase.
  • DNA-dependent RNA polymerases are the T7, T3, and SP6 RNA polymerases.
  • a DNA template for in vitro RNA transcription may be obtained by cloning of a nucleic acid, in particular cDNA corresponding to the respective RNA to be in vitro transcribed, and introducing it into an appropriate vector for in vitro transcription, for example into plasmid DNA.
  • the DNA template is linearized with a suitable restriction enzyme, before it is transcribed in vitro.
  • the cDNA may be obtained by reverse transcription of mRNA or chemical synthesis.
  • the DNA template for in vitro RNA synthesis may also be obtained by gene synthesis.
  • purification/purifying are intended to mean that the target RNA is separated and/or isolated from the by-products and the components of the RNA production (e.g. chemical synthesis, RNA in vitro transcription).
  • the target RNA is separated and/or isolated from the by-products and the components of the RNA in vitro transcription reaction present in the sample comprising the target RNA after the RNA in vitro transcription reaction.
  • the purified target RNA sample has a higher purity than the target RNA-containing sample prior to purification, i.e.
  • RNA in vitro transcription reaction the amount of by-products and the components of the RNA in vitro transcription reaction is lower than in the sample after transcription, but before purification.
  • Undesired constituents of RNA-containing samples which therefore need to be separated may in particular be by-products of the RNA in vitro transcription reaction, or also excessively long transcripts, if plasmids are not completely linearized.
  • components of the RNA in vitro transcription reaction mixture such as enzymes, for example RNases and polymerases, and nucleotides may be separated from the target RNA in the purification step.
  • the target RNA After the purification step, the target RNA has a higher purity than before the purification step, but may still contain by-products which may be detected by the method of the present invention.
  • the degree of purity after the purification step may be more than 70% or 75%, in particular more than 80% or 85%, very particularly more than 90% or 95% and most favorably 99% or more.
  • the degree of purity may for example be determined by an analytical HPLC, wherein the area of the peak for the target RNA is related to the total area of all peaks representing the by-products.
  • Nucleoside standards To be able to identify the compound eluted in any HPLC peak, standards are run under the same conditions, which allows for peak alignment between the sample and the standard, provided that the eluted compound corresponds to one in a standard sample. For the identification of nucleosides or modified nucleosides in an RNA molecule, standards of nucleosides that have been treated in the same way as the sample are run in parallel to the RNA molecule. Furthermore, using different concentrations of each nucleoside standard allows for correlation of the peak area with the amount of nucleoside in the sample.
  • Nucleosides are glycosylamines that correspond to nucleotides without a phosphate group.
  • a nucleoside consists simply of a nucleobase (also termed a nitrogenous base) and a 5-carbon sugar (either ribose or deoxyribose), whereas a nucleotide is composed of a nucleobase, a five-carbon sugar, and one or more phosphate groups.
  • the base is bound to either ribose or deoxyribose via a beta-glycosidic linkage.
  • Examples of nucleosides include cytidine, uridine, adenosine, guanosine, thymidine and inosine.
  • RNA molecule The expected sequence of an RNA molecule is the nucleotide sequence predicted from the sequence of the DNA template from which it is transcribed in the process of RNA in vitro transcription, or the order of synthesis steps in its chemical synthesis protocol, i.e. the nucleotide sequence with which any sequence of an actually produced molecule will be compared during quality control.
  • the measured content of a nucleoside is the actual content of said nucleoside in an in vitro transcribed RNA molecule or chemically synthesized RNA molecule. Within the present invention, the measured content is determined by HPLC analysis of the nucleoside ratios of the hydrolyzed RNA molecules.
  • Expected ratio The expected ratio of a nucleoside is the number of this nucleoside in the expected sequence of an RNA molecule divided by the total number of all nucleosides in the expected sequence.
  • the measured ratio of a nucleoside is the number of this nucleoside detected in in vitro transcribed or chemically synthesized RNA molecules divided by the total number of nucleosides detected in said RNA molecules.
  • the present invention relates to the field of RNA analysis, specifically to methods for quality control in RNA production.
  • the present invention relates to a method for analyzing a sample of RNA molecules transcribed from a template DNA with known sequence and length or chemically synthesized with known sequence and length, comprising the steps of:
  • the method further comprises determining in step d) at least one of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • step d) of the method of the invention may involve the following embodiments:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • the method comprises determining all of:
  • RNA molecules Before completely hydrolyzing the RNA molecules, they may be purified, in particular if the RNA molecules are provided by RNA in vitro transcription. In this case, the purification step removes free nucleotides which were not incorporated into the RNA molecules and which may distort the analysis of the nucleosides released by the hydrolysis step.
  • the RNA molecules may be purified by any suitable method.
  • the method for purifying the RNA molecules is chosen so that the reagents (such as nucleotides and RNA polymerase) and by-products of the RNA in vitro transcription reaction are removed from the sample as completely as possible.
  • Suitable purification methods include alcoholic precipitation, LiCl precipitation, HPLC such as reversed-phase HPLC, anion exchange chromatography, hydroxyapatite chromatography and core bead chromatography, tangential flow filtration, gel filtration chromatography, silica membranes such as, for example, PureYieldTM RNA Midiprep System of Promega and affinity chromatography (if a tag is attached to the target RNA or via oligod(T), if the RNA is polyadenylated).
  • HPLC such as reversed-phase HPLC
  • anion exchange chromatography hydroxyapatite chromatography and core bead chromatography
  • tangential flow filtration gel filtration chromatography
  • silica membranes such as, for example, PureYieldTM RNA Midiprep System of Promega and affinity chromatography (if a tag is attached to the target RNA or via oligod(T), if the RNA is polyadenylated).
  • the HPLC for purifying the RNA is preferably performed on a preparative scale in which relatively large quantities of RNA are purified.
  • Such relatively large quantities are for example quantities of 0.5 mg or more, in particular 1.0 mg to 1000 mg or more, very particularly approximately 1.5 mg or more, upscaling even to the kg range being possible.
  • the above statements are to be understood to mean that these quantities relate to a single HPLC run. If a plurality of HPLC runs is performed, the quantity increases in direct proportion to the number of HPLC runs.
  • a particularly preferred method for purifying the target RNA is disclosed in WO 2008/077592 A1 and involves a reversed-phase HPLC using a porous reversed phase as stationary phase.
  • any material known to be used as reverse phase stationary phase in particular any polymeric material may be used for the inventive method, if that material can be provided in porous form.
  • the stationary phase may be composed of organic and/or inorganic material.
  • polymers to be used for the purification step of the present invention are (non-alkylated) polystyrenes, (non-alkylated) polystyrenedivinylbenzenes, silica gel, silica gel modified with non-polar residues, particularly silica gel modified with alkyl containing residues, more preferably with butyl-, octyl and/or octadecyl containing residues, silica gel modified with phenylic residues, polymethacrylates, etc.
  • the material for the reversed phase is a porous polystyrene polymer, a (non-alkylated) porous polystyrenedivinylbenzene polymer, porous silica gel, porous silica gel modified with non-polar residues, particularly porous silica gel modified with alkyl containing residues, more preferably with butyl-, octyl and/or octadecyl containing residues, porous silica gel modified with phenylic residues, porous polymethacrylates, wherein in particular a porous polystyrene polymer or a non-alkylated (porous) polystyrenedivinylbenzene may be used.
  • a non-alkylated porous polystyrenedivinylbenzene which is very particularly preferred for the purification step of the method according to the invention is one which, without being limited thereto, may have a particle size of 8.0 ⁇ 1.5 ⁇ m, in particular 8.0 ⁇ 0.5 ⁇ m, and a pore size of 1000-1500 ⁇ , in particular 1000-1200 ⁇ or 3500-4500 ⁇ .
  • the stationary phase is conventionally located in a column.
  • V2A steel is conventionally used as the material for the column, but other materials may also be used for the column provided they are suitable for the conditions prevailing during HPLC.
  • the column is straight. It is favourable for the HPLC column to have a length of 5 cm to 100 cm and a diameter of 4 mm to 25 mm.
  • Columns used for the purification step of the method of the invention may in particular have the following dimensions: 50 mm long and 7.5 mm in diameter or 50 mm long and 4.6 mm in diameter, or 50 mm long and 10 mm in diameter or any other dimension with regard to length and diameter, which is suitable for preparative recovery of RNA, even lengths of several meters and also larger diameters being feasible in the case of upscaling.
  • the HPLC is preferably performed as ion-pair, reversed phase HPLC as defined above.
  • a mixture of an aqueous solvent and an organic solvent is used as the mobile phase for eluting the RNA.
  • the buffer used as the aqueous solvent has a pH of 6.0-8.0, for example of about 7, for example 7.0.
  • the buffer is triethylammonium acetate which preferably has a concentration of 0.02 M to 0.5 M, more preferably of 0.08 M to 0.12 M.
  • a 0.1 M triethylammonium acetate buffer is used, which also acts as a counter ion to the RNA in the ion pair method.
  • the organic solvent which is used in the mobile phase is selected from acetonitrile, methanol, ethanol, 1-propanol, 2-propanol and acetone or a mixture thereof. More preferably it is acetonitrile.
  • the mobile phase is a mixture of 0.1 M triethylammonium acetate, pH 7, and acetonitrile.
  • the mobile phase contains 5.0 vol. % to 25.0 vol. % organic solvent, relative to the mobile phase, and for this to be made up to 100 vol. % with the aqueous solvent.
  • the proportion of organic solvent is increased, in particular by at least 10%, more preferably by at least 50% and most preferably by at least 100%, optionally by at least 200%, relative to the initial vol. % in the mobile phase.
  • the proportion of organic solvent in the mobile phase amounts in the course of HPLC separation to 3 to 9, preferably 4 to 7.5, in particular 5.0 vol. %, in each case relative to the mobile phase.
  • the proportion of organic solvent in the mobile phase is increased in the course of HPLC separation from 3 to 9, in particular 5.0 vol. % to up to 20.0 vol. %, in each case relative to the mobile phase. Still more preferably, the method is performed in such a way that the proportion of organic solvent in the mobile phase is increased in the course of HPLC separation from 6.5 to 8.5, in particular 7.5 vol. %, to up to 1 7.5 vol. %, in each case relative to the mobile phase.
  • the mobile phase contains 7.5 vol. % to 17.5 vol. % organic solvent, relative to the mobile phase, and for this to be made up to 100 vol. % with the aqueous buffered solvent.
  • Elution may proceed isocratically or by means of gradient separation.
  • elution of the RNA proceeds with a single eluent or a constant mixture of a plurality of eluents, wherein the solvents described above in detail may be used as eluent.
  • gradient separation is performed wherein the composition of the eluent is varied by means of a gradient program.
  • the equipment necessary for gradient separation is known to a person skilled in the art.
  • Gradient elution may here proceed either on the low pressure side by mixing chambers or on the high pressure side by further pumps.
  • the proportion of organic solvent is increased relative to the aqueous solvent during gradient separation.
  • the above-described agents may here be used as the aqueous solvent and the likewise above-described agents may be used as the organic solvent.
  • the proportion of organic solvent in the mobile phase may be increased in the course of HPLC separation from 5.0 vol. % to 20.0 vol. %, in each case relative to the mobile phase.
  • the proportion of organic solvent in the mobile phase may be increased in the course of HPLC separation from 7.5 vol. % to 1 7.5 vol. %, in particular 9.5 to 14.5 vol. %, in each case relative to the mobile phase.
  • purified solvent for HPLC.
  • Such purified solvents are commercially obtainable. They may additionally also be filtered through a 1 to 5 ⁇ m microfilter, which is generally mounted in the system upstream of the pump. It is additionally preferred for all the solvents to be degassed prior to use, since otherwise gas bubbles occur in most pumps. If air bubbles occur in the solvent, they may interfere not only with separation but also with the continuous monitoring of outflow in the detector.
  • the solvents may be degassed by heating, by vigorous stirring with a magnetic stirrer, by brief evacuation, by ultrasonication or by passing a small stream of helium through the solvent storage vessel.
  • the flow rate of the eluent is selected such that good separation of the RNA from the other constituents contained in the sample to be investigated takes place.
  • the eluent flow rate may amount to from 1 ml/min to several litres per minute (in the case of upscaling), in particular about 1 to 1000 ml/min, more preferably 5 ml to 500 ml/min, even more preferably more than 100 ml/min, depending on the type and scope of the upscaling. This flow rate may be established and regulated by the pump.
  • Detection proceeds preferably with a UV detector at 254 nm, wherein a reference measurement may be made at 600 nm.
  • a reference measurement may be made at 600 nm.
  • any other detection method may alternatively be used, with which the RNA may be detected.
  • RNA-containing eluted solvent quantities For preparative purification of the RNA, it is advisable to collect the RNA-containing eluted solvent quantities. In this respect, it is preferred to carry out this collection in such a way that the eluted solvent is collected in individual separated fractions. This may take place for example with a fraction collector. In this way, the high-purity RNA-containing fractions may be separated from other RNA-containing fractions which still contain undesired impurities, albeit in very small quantities. The individual fractions may be collected for example over 1 minute.
  • the purification by HPLC is preferably performed under completely denaturing conditions. This may proceed for example in that sample application takes place at a temperature of 4-12° C., the HPLC method otherwise proceeding at a higher temperature, preferably at 70° C. or more, particularly preferably at 75° C. or more, in particular up to 82° C., and very particularly preferably at about 78° C.
  • Sample application may be performed with two methods, stop-flow injection or loop injection.
  • stop-flow injection a microsyringe is used which is able to withstand the high pressure applied in HPLC.
  • the sample is injected through a septum in an inlet valve either directly onto the column packing or onto a small drop of inert material immediately over the packing.
  • the system may in this case be under elevated pressure, or the pump may be turned off prior to injection, which is then performed when the pressure has fallen to close to the normal value.
  • a loop injector is used to introduce the sample. This consists of a tubular loop, into which the sample is inserted.
  • the stationary phase is then conveyed out of the pump through the loop, whose outlet leads directly into the column.
  • the sample is entrained in this way by the stationary phase into the column, without solvent flow to the pump being interrupted.
  • the complete hydrolysis of step b) is chemical hydrolysis.
  • the chemical hydrolysis is acid-catalyzed hydrolysis which may be accomplished by treatment of the RNA molecules with trifluoroacetic acid or with HCl at a temperature of 100° C.
  • the chemical hydrolysis is alkaline hydrolysis.
  • the pH of the RNA solution is increased, e.g. to about pH 9, preferably by the addition of an alkaline hydrolysis buffer which may contain sodium carbonate (e.g. 50 mM NaHCO 3 /Na 2 CO 3 , pH 9.2, 1 mM EDTA) and heated to a temperature of about 95° C. for a suitable time.
  • sodium carbonate e.g. 50 mM NaHCO 3 /Na 2 CO 3 , pH 9.2, 1 mM EDTA
  • the complete hydrolysis of step b) is enzymatic hydrolysis.
  • the enzymatic hydrolysis is achieved by treatment with a nuclease, a phosphatase and a phosphodiesterase.
  • the nuclease is Nuclease P1 from Penicillium citrinum .
  • the phosphodiesterase is Phosphodiesterase I from Crotalus adamanteus .
  • the phosphatase is shrimp alkaline phosphatase.
  • the nuclease is Nuclease P1 from Penicillium citrinum and the phosphodiesterase is Phosphodiesterase I from Crotalus adamanteus .
  • the nuclease is Nuclease P1 from Penicillium citrinum and the phosphatase is shrimp alkaline phosphatase.
  • the phosphodiesterase is Phosphodiesterase I from Crotalus adamanteus and the phosphatase is shrimp alkaline phosphatase.
  • the nuclease is Nuclease P1 from Penicillium citrinum
  • the phosphodiesterase is Phosphodiesterase I from Crotalus adamanteus
  • the phosphatase is shrimp alkaline phosphatase.
  • the RNA molecules are first hydrolyzed with nuclease P1, before phosphodiesterase I and shrimp alkaline phosphatase are added to the mixture.
  • the treatment with nuclease P1 is for one to three, preferably for two hours and the treatment with phosphodiesterase I and shrimp alkaline phosphatase takes place for another one to three, preferably for two hours.
  • the temperature for treatment with nuclease P1 is preferably 42° C. and the temperature for the treatment with phosphodiesterase I and shrimp alkaline phosphatase is preferably 37° C.
  • nuclease P1 is used per ⁇ g RNA, more preferably, 0.002 ⁇ g to 0.008 ⁇ g nuclease P1 is used per ⁇ g RNA, even more preferably 0.004 ⁇ g to 0.006 ⁇ g nuclease P1 is used per ⁇ g RNA and most preferably 0.005 ⁇ g nuclease P1 is used per ⁇ g RNA.
  • 0.000005 U to 0.00008 U phosphodiesterase I is used per ⁇ g RNA, more preferably 0.000008 U to 0.00006 U phosphodiesterase I is used per ⁇ g RNA, even more preferably 0.00001 U to 0.00004 U phosphodiesterase I is used per ⁇ g RNA and most preferably 0.0000217 U phosphodiesterase I is used per ⁇ g RNA.
  • 0.05 U to 0.5 U shrimp alkaline phosphatase is used per ⁇ g RNA, more preferably 0.08 U to 0.3 U shrimp alkaline phosphatase is used per ⁇ g RNA, even more preferably 0.1 to 0.2 U shrimp alkaline phosphatase is used per ⁇ g RNA and most preferably 0.167 U shrimp alkaline phosphatase is used per ⁇ g RNA.
  • the sample comprising RNA molecules is first treated with 0.005 ⁇ g nuclease P1 per ⁇ g RNA in a solution containing 33 mM ammonium acetate buffer pH 5.3 supplemented with 2.2 mM ZnCl 2 for two hours at 42° C., and then hydrolyzed with 0.0000217 U phosphodiesterase I per ⁇ g RNA and 0.167 U shrimp alkaline phosphatase per ⁇ g RNA for two more hours at 37° C. in Tris-HCl buffer pH 8.3 with 11.5 mM magnesium acetate.
  • the sample comprises a mixture of all nucleosides present in the RNA molecule which was subjected to hydrolysis.
  • the nucleosides present in the mixture have to be separated from each other so that it is possible to distinguish between the nucleosides.
  • HPLC high-performance capillary electrophoresis
  • MALDI-TOF MS Matrix Assisted Laser Desorption/Ionization-Time of Flight Mass Spectrometry
  • LC-MS Liquid chromatography-mass spectrometry
  • any material known to be used as reverse phase stationary phase in particular any polymeric material may be used, if that material can be provided in porous or monolithic form.
  • the stationary phase may be composed of organic and/or inorganic material.
  • polymers to be used for the purification step of the present invention are (non-alkylated) polystyrenes, (non-alkylated) polystyrenedivinylbenzenes, silica gel, silica gel modified with non-polar residues, particularly silica gel modified with alkyl containing residues, more preferably with butyl-, octyl and/or octadecyl containing residues, silica gel modified with phenylic residues, polymethacrylates, etc.
  • the material for the reversed phase is a porous polystyrene polymer, a (non-alkylated) porous polystyrenedivinylbenzene polymer, porous silica gel, porous silica gel modified with non-polar residues, particularly porous silica gel modified with alkyl containing residues, more preferably with butyl-, octyl and/or octadecyl containing residues, porous silica gel modified with phenylic residues, porous polymethacrylates, wherein in particular a porous polystyrene polymer or a non-alkylated (porous) polystyrenedivinylbenzene may be used.
  • An octadecyl capped, porous silica gel column which is very particularly preferred for the separation and detection of the nucleosides is one which, without being limited thereto, may have a particle size of 10-50 ⁇ m, particularly 10 ⁇ m and a pore size of 120-300 ⁇ , particularly 300 ⁇ .
  • the stationary phase is conventionally located in a column.
  • V2A steel is conventionally used as the material for the column, but other materials may also be used for the column provided they are suitable for the conditions prevailing during HPLC.
  • the column is straight. It is favorable for the HPLC column to have a length of 5 cm to 100 cm and a diameter of 4 mm to 25 mm.
  • Columns used for step c) of the method of the invention may in particular have the following dimensions: 250 mm long and 4.6 mm in diameter.
  • the nucleosides are applied to the HPLC column in an aqueous solvent, preferably a buffer.
  • the buffer used as the aqueous solvent has a pH of 5.0-7.0, more preferably of 5.5 to 6.5 and most preferably of 6.0.
  • the buffer is ammonium acetate which preferably has a concentration of 0.001 M to 0.01 M, more preferably of 0.002 M to 0.007 M.
  • a 0.005 M ammonium acetate buffer pH 6.0 is used.
  • the organic solvent which is used for eluting the nucleosides is selected from acetonitrile, methanol, ethanol, 1-propanol, 2-propanol and acetone or a mixture thereof. More preferably, the organic solvent is acetonitrile. Most preferably, it is 40 vol. % acetonitrile.
  • Elution may proceed isocratically or by means of gradient separation.
  • elution of the RNA proceeds with a single eluent or a constant mixture of a plurality of eluents, wherein the solvents described above in detail may be used as eluent.
  • gradient separation is performed wherein the composition of the eluent is varied by means of a gradient program.
  • the equipment necessary for gradient separation is known to a person skilled in the art.
  • Gradient elution may here proceed either on the low pressure side by mixing chambers or on the high pressure side by further pumps.
  • the proportion of organic solvent is increased relative to the aqueous solvent during gradient separation.
  • the above-described agents may here be used as the aqueous solvent and the likewise above-described agents may be used as the organic solvent.
  • the proportion of organic solvent in the mobile phase preferably of acetonitrile, may be increased in the course of HPLC separation from 0.0 vol. % to 20.0 vol. %, in each case relative to the mobile phase.
  • purified solvent for HPLC.
  • Such purified solvents are commercially obtainable. They may additionally also be filtered through a 1 to 5 ⁇ m microfilter, which is generally mounted in the system upstream of the pump. It is additionally preferred for all the solvents to be degassed prior to use, since otherwise gas bubbles occur in most pumps. If air bubbles occur in the solvent, they may interfere not only with separation but also with the continuous monitoring of outflow in the detector.
  • the solvents may be degassed by heating, by vigorous stirring with a magnetic stirrer, by brief evacuation, by ultrasonication or by passing a small stream of helium through the solvent storage vessel.
  • the flow rate of the eluent is selected such that good separation of the nucleosides contained in the sample to be investigated takes place.
  • the eluent flow rate may amount to from 0.5 ml/min to 5 milliliters per minute, in particular about 0.6 to 2 ml/min, more preferably 0.7 ml to 10 ml/min, even more preferably 0.85 ml/min. This flow rate may be established and regulated by the pump.
  • Detection proceeds preferably with a UV detector at 254 nm, wherein a reference measurement may be made at 600 nm.
  • a reference measurement may be made at 600 nm.
  • any other detection method may alternatively be used, with which the nucleosides may be detected.
  • Sample application may be performed with two methods, stop-flow injection or loop injection.
  • stop-flow injection a microsyringe is used which is able to withstand the high pressure applied in HPLC.
  • the sample is injected through a septum in an inlet valve either directly onto the column packing or onto a small drop of inert material immediately over the packing.
  • the system may in this case be under elevated pressure, or the pump may be turned off prior to injection, which is then performed when the pressure has fallen to close to the normal value.
  • a loop injector is used to introduce the sample. This consists of a tubular loop, into which the sample is inserted.
  • the stationary phase is then conveyed out of the pump through the loop, whose outlet leads directly into the column.
  • the sample is entrained in this way by the stationary phase into the column, without solvent flow to the pump being interrupted.
  • detecting the released nucleosides comprises identifying the released nucleosides by comparison to a standard for each nucleoside. In an embodiment, this standard is obtained by completely hydrolyzing the corresponding nucleoside triphosphate and detecting the resulting nucleosides by HPLC.
  • the corresponding nucleoside triphosphate is treated in the same way as the sample comprising RNA molecules, i.e. if the sample comprising RNA molecules is enzymatically hydrolyzed as described above, the corresponding nucleoside triphosphate is treated with the same enzymes and under the same conditions. If the sample comprising RNA molecules is subjected to chemical hydrolysis, the corresponding nucleoside triphosphate is also subjected to chemical hydrolysis.
  • step c) of the inventive method further comprises a step c1) of quantifying the released nucleosides.
  • the released nucleosides are quantified by determining the peak area for each of the nucleosides and using the linear standard plot method (Li et al. (2012) Fitorick 83:1267-1274). For this purpose, a linear standard curve is created by completely hydrolyzing nucleotides into nucleosides, diluting 500 ⁇ M of the relevant nucleoside 1:1 ten times, subjecting these dilutions to HPLC analysis as described above, and plotting the values of the peak areas against the concentration to create a linear standard curve.
  • the obtained number will be the amount of RNA molecules in the sample.
  • A is omitted from the formula since polyadenylation (addition of homopolymeric A-nucleotide element) would skew the calculation toward a higher number.
  • step d) of the inventive method comprises determining the matching of the sequence of the RNA molecules with that of the known sequence and/or the matching of the length of the RNA molecules with the known length by comparing the measured ratio of a specific nucleoside in the sample (rm) with the expected ratio of said nucleoside (re).
  • the measured and expected ratio is calculated for each of C, G, and U in polyadenylated RNA molecules.
  • RNA molecules comprising homopolymeric elements other than a poly(A) tail (for example a poly(C) stretch) the measured and expected ratio is calculated for each of A, G, and U in poly(C) stretch containing RNA molecules.
  • the obtained values for rm(X) and re(X) can then be compared.
  • the values of the measured and expected ratio for the same nucleoside are compared.
  • a close match between the measured and expected ratio indicates that both the sequence and the length of the RNA molecules closely match with the expected sequence and length.
  • a low match between the measured and expected ratio could indicate that the sequence of the RNA molecules does not match with the expected sequence, e.g. that the produced RNA molecules are not the desired ones, or that transcription errors occurred, or that the RNA molecules have a different length as the expected length, e.g. that transcription did not proceed to completion or that the RNA was (partially) degraded after transcription.
  • the ratio is calculated for each of C, G, U and A in a non-polyadenylated RNA molecule.
  • rm(X) and re(X) can then be compared.
  • the values of the measured and expected ratio for the same nucleoside are compared.
  • a low match between the measured and expected ratio could indicate that the sequence of the RNA molecules does not match with the expected sequence, e.g. that the produced RNA molecules are not the desired ones, or that transcription errors occurred, or that the length of the RNA molecules does not match with the expected length, e.g. that transcription did not proceed to completion or that the RNA was (partially) degraded after transcription.
  • a high % cap value indicates that a high percentage of RNA molecules within the sample contain a cap structure.
  • the cap analogue is selected from the group consisting of:
  • the number of copies of the RNA molecules in the sample is determined using formula 1.1 if the RNA molecules are polyadenylated, or using formula 1.2 if the RNA molecules are not polyadenylated.
  • the RNA is capped using enzymatic capping.
  • the number of copies of the RNA molecules in the sample may be determined by using formula 1.1.
  • the modified nucleoside is selected from the group of: 2-amino-6-chloropurineriboside-5′-triphosphate, 2-Aminopurine-riboside-5′-triphosphate; 2-aminoadenosine-5′-triphosphate, 2′-Amino-2′-deoxycytidine-triphosphate, 2-thiocytidine-5′-triphosphate, 2-thiouridine-5′-triphosphate, 2′-Fluorothymidine-5′-triphosphate, 2′-O-Methyl inosine-5′-triphosphate 4-thiouridine-5′-triphosphate, 5-aminoallylcytidine-5′-triphosphate, 5-aminoallyluridine-5′-triphosphate, 5-bromocytidine-5′-triphosphate, 5-bromouridine-5′-triphosphate, 5-Bromo-2′-deoxycytidine-5′-triphosphate, 5-Bromo-2′-deoxyuridine, 5-B
  • the modified nucleosides are oxidized nucleosides so that the percentage of their incorporation into the RNA molecules allows to determine the oxidation status of the RNA molecules.
  • the oxidized nucleosides are preferably selected from the group consisting of: 8-hydroxyguanosine (8-OHG), 8-oxo-7,8-dihydro-2′-deoxyguanosine, dehydroguanidinohyantoin, 2,4,6-trioxo-[1,3,5]triazinane-1-carboxamidine, N-nitro-dehydroguanidinohydantoin, spiroiminodihydantoin, guanidinohydantoin, 4-hydroxy-2,5-dioxo-imidazolidine-4-carboxylic acid, 3-nitrotyrosine, 3.3′-dityrosine, 3-hydroxy-5-imino-3,3a,4,5-tetrahydro-1H-imidazo[4,5-d]
  • the RNA molecules to be analyzed is selected from the group consisting of: messenger RNA (mRNA), a viral RNA, retroviral RNA and replicon RNA, small interfering RNA (siRNA), antisense RNA (asRNA), circular RNA(circRNA), CRISPR/Cas9 guide RNA, ribozymes, aptamers, riboswitches, immunostimulating RNA, transfer RNA (tRNA), ribosomal RNA (rRNA), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), microRNA (miRNA), and Piwi-interacting RNA (piRNA).
  • mRNA messenger RNA
  • a viral RNA retroviral RNA and replicon RNA
  • siRNA small interfering RNA
  • asRNA antisense RNA
  • circular RNA(circRNA) circular RNA(circRNA)
  • CRISPR/Cas9 guide RNA CRISPR/Cas9 guide
  • the RNA molecules being analyzed are mRNA molecules.
  • the inventive method additionally comprises a step a.1) of modifying the RNA molecules.
  • modifying the RNA molecules comprises enzymatically adding a poly(A) tail to the RNA molecules. This is achieved by reacting the RNA molecules with poly(A)polymerase using 1 mM ATP at 37° C. for 5 to 120 minutes, depending on the desired poly (A) tail length, preferably for 30 to 60 minutes.
  • the poly(A)polymerase is derived from Escherichia coli, Streptomyces coelicolor, Meiothermus silvanus, Bacillus subtilis, Thermus aquaticus, Shigella flexneri, Shigella dysenteriae, Citrobacter koseri, Salmonella bongori, Salmonella enterica, Trabulsiella guamensis, Kluyvera ascorbata, Citrobacter freundii, Enterobacter cloacae, Enterococcus gallinarum, Grimontia indica , or Salinivibrio costicola , most preferably from Escherichia coli.
  • the number of copies of the RNA molecules in the sample is determined using formula 1.1.
  • modifying the RNA molecules comprises enzymatically adding a cap structure to the RNA molecules.
  • Enzymatic capping occurs post-transcriptionally and uses a eukaryotic or viral capping enzyme, more preferably a Vaccinia virus caping enzyme (to generate cap0), still more preferably a Vaccinia virus capping enzyme and a Vaccinia virus cap-specific nucleoside 2′-O-methyltransferase (to generate cap1).
  • the RNA molecules are reacted with the capping enzyme, a nucleotide, GTP, and a methyl donor, preferably S-adenosylmethionine.
  • the capping reaction occurs at 25° C. for 1 hour, using for example 0.5 mg/ml RNA, 5 M nucleotide, GTP, and 1 M methyl donor, preferably S-adenosylmethionine.
  • modifying the RNA molecules comprises enzymatically adding a cap1 structure to an RNA comprising a cap0 structure.
  • Vaccinia virus cap-specific nucleoside 2′-O-methyltransferase is used under suitable conditions (as explained above).
  • the number of copies of the RNA molecules in the sample is determined using formula 1.1, if the RNA molecules are polyadenylated, or using formula 1.2, if the RNA molecules are not polyadenylated.
  • the inventive method is used in the quality control of RNA prepared by RNA in vitro transcription.
  • the inventive method is used in the quality control of RNA prepared by chemical synthesis.
  • Such a quality control involves determining the number of copies, matching of the sequence with the expected sequence, matching of the length with the expected length, purity and other aspects of the RNA molecules, and is particularly important in the production of RNA which is to be used for administration to humans or animals, for example for therapeutic purposes.
  • the sample comprising RNA molecules shows the required quality in terms of number of copies, matching of the sequence with the expected sequence, matching of the length with the expected length, and homopolymeric elements, in the case of mRNA molecules in terms of number of copies, matching of the sequence with the expected sequence, matching of the length with the expected length, poly(A) tail, and capping, and optionally for RNA or mRNA molecules incorporation of modified nucleotides and oxidation status
  • the sample can be processed further to be ultimately administered to a human or animal subject. If any of the criteria above are not met, the sample comprising RNA molecules will be discarded, as it is not suitable for administration to humans or animals.
  • DNA sequences encoding mRNA of Photinus pyralis luciferase (PpLuc, R2988), three human cancer antigens (HsPSCA, R1871; HsNY-ESO-1, R1857; Hs5T4, R1855) and Rabies virus glycoprotein (RAV-G, R1803) were generated.
  • the DNA sequences were prepared by modifying the wild type encoding DNA sequence to optimize the GC-content for stabilization.
  • the DNA sequence was introduced into a pUC19 derived vector and modified to comprise an alpha-globin-3′-UTR (muag (mutated alpha-globin-3′-UTR)), a histone-stem-loop structure, and a stretch of 70 ⁇ adenosine at the 3′-terminal end (poly-A-tail).
  • the obtained plasmid DNA was used for RNA in vitro transcription experiments to obtain respective mRNA (SEQ ID NO. 1-5).
  • the DNA plasmids prepared according to paragraph 1 were transcribed in vitro using T7 polymerase. All mRNAs were capped by co-transcriptional capping, using a cap analogue. In addition to that, for the preparation of PpLuc mRNA (SEQ ID No. 1), a modified nucleotide 8-OH-G was incorporated (detailed description see below).
  • Linearized plasmid DNA template (50 ⁇ g/ml) was transcribed at 37° C. for 3-5 hours in 80 mM HEPES/KOH, pH 7.5, 24 mM MgCl 2 , 2 mM spermidine, 40 mM DTT, 5 U/ml pyrophosphatase (Thermo Fisher Scientific), 200 U/ml RiboLock RNase inhibitor (Thermo Fisher Scientific), 5000 U/ml T7 RNA polymerase (Thermo Fisher Scientific).
  • PpLuc (8-OH-G) an PpLuc mRNA was synthetized comprising the modified nucleotide 8-OH-G.
  • PpLuc (8-OH-G) 25% of the amount of GTP applied in the RNA in vitro transcription reaction was replaced with 8-OH-GTP.
  • the respective NTP composition used for the in vitro transcription of PpLuc (8-OH-G) was 1.025 mM 8-OH-GTP, 3.075 mM GTP, 4.36 mM CTP, 3.07 mM ATP, 1.92 mM UTP, 16.4 mM m7G(5′)ppp(5′)G Cap.
  • RNAs were precipitated in 2.86 M LiCl for 16 hours at ⁇ 20° C., followed by centrifugation (30 min, 16.000 g, 4° C.). Pellets were washed in 0.1 transcription reaction volumes of 75% ethanol (invert, centrifuge 5 min, 16.000 g, 4° C.), dried and re-dissolved in 10 transcription reaction volumes H 2 O.
  • mRNA was purified using PureMessenger® (see WO 2008/077592A1).
  • PpLuc (8-OH-G) mRNA was purified with RNeasy columns (Qiagen) according to the manufacturer's instructions.
  • the expected sequences are provided (SEQ ID NO. 1-5).
  • the expected nucleotide composition (A,G,U,C) of each synthetized mRNA is indicated (see Table 1).
  • the goal of this experiment was to enzymatically hydrolyze mRNA species obtained according to Example 1 to obtain the single nucleosides of each of the mRNA species.
  • the obtained nucleoside mixtures were used for further analysis using HPLC (see Example 3).
  • mRNA was completely hydrolysed by the action of three types of hydrolases.
  • Phosphodiesterase I from Crotalus adamanteus (SvP) and Nuclease P1 from Penicillium citrinum (P1) were obtained as dried powder from Sigma Aldrich.
  • SvP was dissolved to 0.5 U/ml in 110 mM Tris-HCl buffer, pH 8.9, 100 mM NaCl, 15 mM MgCl2, 50% Glycerol and stored at ⁇ 20° C.
  • P1 was dissolved to 1 g/L in 20 mM sodium acetate buffer pH 5.3, 5 mM ZnCl2, 50 mM NaCl, 50% Glycerol and stored at ⁇ 20° C.
  • Shrimp alkaline phosphatase (AP) was obtained from New England Biolabs.
  • each mRNA was hydrolysed first for 2 h at 42° C. with 0.3 ⁇ g of P1 in 33 mM ammonium acetate buffer pH 5.3 supplemented with 2.2 mM ZnCl 2 .
  • Tris-HCl buffer (pH 8.3), magnesium acetate, SvP and AP were added to 11.5 mM, 1.15 mM, 10 U/1 or 0.077 U/ ⁇ 1, respectively (reaction volume: 130 ⁇ l).
  • the hydrolysis was allowed to proceed for 2 h at 37° C. After that, the reaction was centrifuged for 10 min at 30,000 g. The supernatant was supplemented with 2.2 ⁇ l of ammonium acetate buffer pH 5.3 (300 mM). 20 ⁇ l of the resulting solution were analyzed by HPLC (see Example 3)
  • the goal of this experiment was to analyse each nucleoside mixture obtained via total mRNA hydrolysis (see Example 2) by HPLC to gain information about the matching of the sequence with the expected sequence, the matching of the length with the expected length, the capping efficiency, the incorporation rate of modified nucleosides (8-OH-G), oxidation and the length of the poly(A) stretch of each hydrolysed mRNA.
  • This inventive method can be used as a multifaceted quality control of in vitro transcribed mRNA.
  • Example 2 Samples obtained via total hydrolysis (see Example 2) were separated using a commercially available HPLC setup.
  • an octadecyl capped silica column (YMC*Gel ODS-A-HG, pore size: 300 A, particle size: 10 ⁇ m, dimensions (ID ⁇ h) 250 mm ⁇ 4.6 mm, YMC) in a linear gradient from 100% Buffer A (5 mM NH 4 OAc pH 6) to 20% Buffer B (40% v/v acetonitrile) was used for the separation of nucleosides in the hydrolysate samples.
  • nucleotide triphosphates were hydrolysed (according to Example 2) and analysed at 254 nm to obtain standards for calibration.
  • a commercially available HPLC machine was used for the separation of hydrolysates and nucleoside standards.
  • the nucleosides were detected at a wavelength of 254 nm.
  • Exemplary chromatograms for a hydrolysate of PpLuc mRNA and respective standards are shown in FIG. 1 .
  • the expected content of C, G, A, U, m7G (cap analogue) and optionally 8-OH-G in each mRNA hydrolysate sample was determined (n(C), n(G), n(A), n(U), n(m7G), n(8-OH-G)).
  • the number of C, G, A, and U in each mRNA sequence was determined (u(C), u(G), u(A), u(U)) (see Table 1).
  • n (RNA) 1 ⁇ 3*( n ( C )/ u ( C )+[ n ( G )+ n (8- OH - G )]/ u ( G )+ u (8- OH - G )]+ n ( U )/ u ( U )).
  • the obtained mRNA hydrolysates were analyzed for their nucleoside composition.
  • This measurement can be used to reliably identify mRNA species because the measured ratio of C:G:U has to match the expected ratio of C:G:U.
  • This measurement is particularly useful as a quality control step of in vitro transcribed mRNA for the determination of mRNA identity, i.e. whether the sequence of the RNA molecules matches with the expected sequence.
  • the expected C:G:U values do not match the measured C:G:U values, it will indicate that the mRNA has a length which differs from the expected length.
  • the measured ratio of nucleosides of one mRNA species was compared to the expected ratios of said mRNA species.
  • FIG. 2 illustrates that the inventive method is particularly suitable for measuring mRNA quality attributes such as mRNA identity.
  • the measured G:C:U ratios and the expected G:C:U ratios largely match, which shows that the method allows a robust identification of mRNA species.
  • this could be an indication for a difference in the length of the RNAs.
  • abortive sequences that could potentially be produced during the process of RNA in vitro transcription may bias the measured G:C:U ratio.
  • n(CA) The amount of m7G (cap analogue) from the capping structure in the mRNA hydrolysate was determined (n(CA)).
  • the 5′ cap of mRNA is an essential structure of protein coding mRNAs, because non-capped mRNA is not translated into protein. Therefore, determining the capping efficiency of in vitro transcribed mRNA is a key quality control of in vitro transcribed mRNA.
  • FIG. 3 illustrates that the inventive method is particularly suitable for measuring mRNA quality attributes such as mRNA capping efficiency.
  • Capping is a key feature of mRNA because non-capped mRNA is not translated into proteins.
  • the content of adenosines of an mRNA sample can be used to determine the length of a poly A-stretch present in the respective mRNA species (expected value: AXe). This measurement is particularly useful as a quality control step of in vitro transcribed mRNA, because plasmid encoded Poly(A) stretches can vary. A similar measurement can also be applied for mRNAs with enzymatically added Poly(A)tails (see Example 4).
  • FIG. 4 illustrates that the inventive method is also suitable for measuring mRNA quality attributes such as the length of the Poly(A) stretch. It is also possible to adapt the measurement in a way that other homopolymeric elements in an mRNA (e.g., a Poly(C) region) can be analyzed.
  • mRNA quality attributes such as the length of the Poly(A) stretch.
  • other homopolymeric elements in an mRNA e.g., a Poly(C) region
  • 8-OH-G was incorporated into the mRNA of PpLuc by replacing 25% of GTP with 8-OH-GTP during RNA in vitro transcription, yielding PpLuc(8-OH-G) (see Example 1).
  • FIG. 5 illustrates that the inventive method is also suitable for determining the incorporation and amount of modified nucleotides.
  • 8-OH-G is an indicator for mRNA oxidation. Therefore, the inventive method is also suitable for determining whether the produced mRNA is oxidized.
  • This measurement is particularly useful as a quality control of in vitro transcribed mRNA, because 8-OH-G is an indicator of mRNA oxidation. Moreover, this measurement can also be used and adapted for the detection of any modified nucleotide that may be incorporated into an mRNA, and is therefore particularly important as a quality control in the production process of mRNA containing modified nucleotides.
  • the goal of this experiment is to use the HPLC method according to Example 3 to assess the length of a poly(A) tail that has been enzymatically added. This is a crucial quality control of mRNA. In addition to that, the capping efficiency is characterized and the nucleoside composition is analyzed.
  • mRNA obtained from RNA in vitro transcription according to Example 1 was polyadenylated using a conventional polyadenylation kit according to the manufacturers' instructions (Poly(A) Polymerase Tailing Kit; epicenter).
  • mRNA is reacted with Escherichia coli poly(A) polymerase using 1 mM ATP at 37° C. for 30 or 60 min (dependent on the desired poly-A tail length).
  • the mRNA is purified using PureMessenger® as described in WO 2008/077592A1.
  • the polyadenylated and purified mRNA is optionally run on a gel to roughly assess mRNA extension.
  • the polyadenylated mRNA is first hydrolyzed (according to Example 2) and subsequently analyzed via HPLC (according to Example 3).
  • both poly(A) tail length and capping efficiency are calculated.
  • the nucleoside composition is determined for measurement of the matching of the length with the expected length and the matching of the sequence with the expected sequence.
US16/081,863 2016-03-03 2017-03-03 RNA analysis by total hydrolysis and quantification of released nucleosides Active 2041-05-04 US11920174B2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EPPCT/EP2016/054599 2016-03-03
EP2016054599 2016-03-03
WOPCT/EP2016/054599 2016-03-03
PCT/EP2017/055048 WO2017149139A1 (fr) 2016-03-03 2017-03-03 Analyse d'arn par hydrolyse totale

Publications (2)

Publication Number Publication Date
US20190100784A1 US20190100784A1 (en) 2019-04-04
US11920174B2 true US11920174B2 (en) 2024-03-05

Family

ID=55587245

Family Applications (1)

Application Number Title Priority Date Filing Date
US16/081,863 Active 2041-05-04 US11920174B2 (en) 2016-03-03 2017-03-03 RNA analysis by total hydrolysis and quantification of released nucleosides

Country Status (3)

Country Link
US (1) US11920174B2 (fr)
EP (1) EP3423595A1 (fr)
WO (1) WO2017149139A1 (fr)

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2014220957A1 (en) 2013-02-22 2015-07-30 Curevac Ag Combination of vaccination and inhibition of the PD-1 pathway
EP2971098B1 (fr) 2013-03-14 2018-11-21 Translate Bio, Inc. Évaluation quantitative de l'efficacité de coiffage de l'arn messager
EP2971102B8 (fr) 2013-03-14 2018-08-15 Translate Bio, Inc. Évaluation quantitative pour l'efficacité de coiffage de l'arn messager
TR201901310T4 (tr) 2013-03-14 2019-02-21 Translate Bio Inc Mesajcı RNA'nın saflaştırılması yöntemleri.
SG10201801431TA (en) 2013-08-21 2018-04-27 Curevac Ag Respiratory syncytial virus (rsv) vaccine
CA2944800A1 (fr) 2014-04-25 2015-10-29 Shire Human Genetic Therapies, Inc. Procedes de purification de l'arn messager
US11149278B2 (en) 2014-12-12 2021-10-19 Curevac Ag Artificial nucleic acid molecules for improved protein expression
DK3294885T3 (da) 2015-05-08 2020-08-10 Curevac Real Estate Gmbh Fremgangsmåde til at fremstille rna
US10760070B2 (en) 2015-05-29 2020-09-01 Curevac Real Estate Gmbh Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
EP3362576A1 (fr) 2015-10-12 2018-08-22 CureVac AG Procédé automatisé d'isolement, de sélection et/ou de détection de micro-organismes ou de cellules présents dans une solution
EP3373965A1 (fr) 2015-11-09 2018-09-19 CureVac AG Vaccins contre les rotavirus
DK3319622T3 (da) 2015-12-22 2020-05-04 Curevac Ag Fremgangsmåde til fremstilling af rna-molekylesammensætninger
US11248223B2 (en) 2015-12-23 2022-02-15 Curevac Ag Method of RNA in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
US11723967B2 (en) 2016-02-17 2023-08-15 CureVac SE Zika virus vaccine
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
US11596699B2 (en) 2016-04-29 2023-03-07 CureVac SE RNA encoding an antibody
EP3452493A1 (fr) 2016-05-04 2019-03-13 CureVac AG Molécules d'acide nucléique et leurs utilisations
EP3452101A2 (fr) 2016-05-04 2019-03-13 CureVac AG Arn codant pour une protéine thérapeutique
US11478552B2 (en) 2016-06-09 2022-10-25 Curevac Ag Hybrid carriers for nucleic acid cargo
US11279923B2 (en) 2016-11-28 2022-03-22 Curevac Ag Method for purifying RNA
WO2018104540A1 (fr) 2016-12-08 2018-06-14 Curevac Ag Arn pour la cicatrisation des plaies
CN110582304A (zh) 2016-12-08 2019-12-17 库尔维科公司 用于治疗或预防肝脏疾病的rna
WO2018115527A2 (fr) 2016-12-23 2018-06-28 Curevac Ag Vaccin contre le coronavirus du syndrome respiratoire du moyen-orient
WO2018115507A2 (fr) 2016-12-23 2018-06-28 Curevac Ag Vaccin contre l'hénipavirus
WO2018115525A1 (fr) 2016-12-23 2018-06-28 Curevac Ag Vaccin contre le virus de lassa
SG10202110491PA (en) 2017-03-24 2021-11-29 Curevac Ag Nucleic acids encoding crispr-associated proteins and uses thereof
WO2018191657A1 (fr) 2017-04-13 2018-10-18 Acuitas Therapeutics, Inc. Lipides pour administration d'agents actifs
JP2020530980A (ja) 2017-07-04 2020-11-05 キュアバック アーゲー 新規核酸分子
US11602557B2 (en) 2017-08-22 2023-03-14 Cure Vac SE Bunyavirales vaccine
EP3701018A4 (fr) 2017-10-27 2021-08-04 University of Cincinnati Compositions, procédés et kits pour la digestion en une étape d'acide nucléique pour analyse par chromatographie en phase liquide-spectrométrie de masse en tandem
EP3707271A1 (fr) 2017-11-08 2020-09-16 CureVac AG Adaptation de séquence d'arn
WO2019115635A1 (fr) 2017-12-13 2019-06-20 Curevac Ag Vaccin contre les flavivirus
US11525158B2 (en) 2017-12-21 2022-12-13 CureVac SE Linear double stranded DNA coupled to a single support or a tag and methods for producing said linear double stranded DNA
EP3802821A4 (fr) * 2018-05-25 2022-06-08 New York Institute of Technology Procédé de séquençage direct d'acides nucléiques
CA3108544A1 (fr) 2018-08-24 2020-02-27 Translate Bio, Inc. Procedes de purification d'arn messager
JP6660994B1 (ja) * 2018-11-22 2020-03-11 フォーデイズ株式会社 核酸(dna及びrna)由来のヌクレオチド、ヌクレオシド及び/又は塩基の定量方法
SG11202106666UA (en) 2018-12-21 2021-07-29 Curevac Ag Methods for rna analysis
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
US11241493B2 (en) 2020-02-04 2022-02-08 Curevac Ag Coronavirus vaccine
EP4165205A1 (fr) * 2020-06-15 2023-04-19 CureVac SE Analyse de mélanges d'acides nucléiques
EP4267178A1 (fr) 2020-12-22 2023-11-01 CureVac SE Vaccin à arn contre des variants sras-cov-2

Citations (147)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002098443A2 (fr) 2001-06-05 2002-12-12 Curevac Gmbh Composition pharmaceutique contenant un arnm stabilise et optimise pour la traduction dans ses regions codantes
US20050059624A1 (en) 2001-12-19 2005-03-17 Ingmar Hoerr Application of mRNA for use as a therapeutic against tumour diseases
EP1518928A1 (fr) 2003-09-26 2005-03-30 Fundacion Pablo Cassara Oligonucléotides à activité antiandrogène pour le traitement des désordres cutanés associés aux androgènes. Compositions pharmaceutiques et leur utilisation
US20050250723A1 (en) 2002-07-03 2005-11-10 Ingmar Hoerr Immunostimulation by chemically modified RNA
US7074596B2 (en) 2002-03-25 2006-07-11 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Synthesis and use of anti-reverse mRNA cap analogues
US20060188490A1 (en) 2003-08-05 2006-08-24 Ingmar Hoerr Transfection of blood cells with mRNA for immune stimulation and gene therapy
US20060275747A1 (en) 2001-12-07 2006-12-07 Hardy Stephen F Endogenous retrovirus up-regulated in prostate cancer
US20080025944A1 (en) 2004-09-02 2008-01-31 Cure Vac Gmbh Combination Therapy for Immunostimulation
WO2008016473A2 (fr) 2006-07-28 2008-02-07 Applera Corporation ANALOGUES DE COIFFES DE NUCLÉOTIDE D'ARNm
WO2008077592A1 (fr) 2006-12-22 2008-07-03 Curevac Gmbh Procédé de purification d'arn à l'échelle préparative par hplc
US20080267873A1 (en) 2005-05-19 2008-10-30 Curevac Gmbh Injection Solution for Rna
WO2008157688A2 (fr) 2007-06-19 2008-12-24 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Synthèse et utilisation d'analogues de phosphorothioate anti-inverse de la coiffe d'arn messager
WO2009149253A2 (fr) 2008-06-06 2009-12-10 Uniwersytet Warszawski Analogues d'arnm cap
US20090324584A1 (en) 2006-07-31 2009-12-31 Curevac Gmbh Nucleic Acid of Formula (I): GIXmGn, or (II): CIXmCn, in Particular as an Immune-Stimulating Agent/Adjuvant
US20100189729A1 (en) 2007-01-09 2010-07-29 Curvac Gmbh Rna-coded antibody
US20100203076A1 (en) 2007-09-04 2010-08-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
WO2010093820A2 (fr) 2009-02-11 2010-08-19 Orion Genomics Llc Combinaisons de polymorphismes destinées à déterminer l'expression spécifique des allèles d'igf2
WO2010123501A1 (fr) 2009-04-22 2010-10-28 Massachusetts Institute Of Technology Suppression immunitaire innée permettant la distribution répétée de longues molécules d'arn
US20100291156A1 (en) 2007-10-09 2010-11-18 Marijke Barner Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US20100305196A1 (en) 2007-10-09 2010-12-02 Cure Vac Gmbh COMPOSITION FOR TREATING PROSTATE CANCER (PCa)
WO2011015347A1 (fr) 2009-08-05 2011-02-10 Biontech Ag Composition vaccinale contenant de l'arn dont la coiffe en 5' est modifiée
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
US20110250225A1 (en) 2008-09-30 2011-10-13 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
US20120021043A1 (en) 2008-01-31 2012-01-26 CureVac GmbHÛ Nucleic Acids Comprising formula (NuGlXmGnNv)a and Derivatives Thereof as Immunostimulating Agent/Adjuvant
WO2012019630A1 (fr) 2010-08-13 2012-02-16 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'une protéine codée
WO2012104360A1 (fr) 2011-02-03 2012-08-09 University Of Veterinary Medicine Vienna Test rt-pcr pour la détection du virus de l'hépatite e aviaire
WO2012135805A2 (fr) 2011-03-31 2012-10-04 modeRNA Therapeutics Administration et formulation d'acides nucléiques génétiquement modifiés
US20120258046A1 (en) 2009-12-09 2012-10-11 Thorsten Mutzke Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
WO2013049231A1 (fr) 2011-09-28 2013-04-04 Htg Molecular Diagnostics, Inc. Procédés de co-détection d'arnm et d'un petit arn non codant
WO2013052523A1 (fr) 2011-10-03 2013-04-11 modeRNA Therapeutics Nucléosides, nucléotides et acides nucléiques modifiés, et leurs utilisations
WO2013059475A1 (fr) 2011-10-18 2013-04-25 Life Technologies Corporation Analogues de coiffes à dérivation alcynyle, préparation et utilisations associées
US20130142818A1 (en) 2010-07-30 2013-06-06 Curevac Gmbh Complexation of nucleic acids with disulfide-crosslinked cationic components for transfection and immunostimulation
WO2013143700A2 (fr) 2012-03-27 2013-10-03 Curevac Gmbh Molécules d'acide nucléique artificielles comprenant une 5'top utr
US20130259879A1 (en) 2011-02-21 2013-10-03 Curevac Gmbh Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
US20130280283A1 (en) 2010-12-29 2013-10-24 Curevac Gmbh Combination of vaccination and inhibition of mhc class i restricted antigen presentation
US20130295043A1 (en) 2011-03-02 2013-11-07 Curevac Gmbh Vaccination in elderly patients
US20130336998A1 (en) 2011-03-02 2013-12-19 Curevac Gmbh Vaccination in newborns and infants
WO2014005038A1 (fr) 2012-06-29 2014-01-03 Htg Molecular Diagnostics, Inc. Procédés de protection d'une nucléase pour la détection de variants nucléotidiques
EP2742951A2 (fr) 2004-09-14 2014-06-18 Argos Therapeutics, Inc. Amplification de souches indépendantes d'polynucleotides de VIH
WO2014152659A1 (fr) 2013-03-14 2014-09-25 Shire Human Genetic Therapies, Inc. Évaluation quantitative pour l'efficacité de coiffage de l'arn messager
WO2015001416A1 (fr) 2013-07-05 2015-01-08 Tata Consultancy Services Limited Regroupement de données multidimensionnelles
US20150037326A1 (en) 2012-01-31 2015-02-05 Bayer Innovation Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and an antigen
US20150057340A1 (en) 2012-02-15 2015-02-26 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
US20150093413A1 (en) 2012-02-15 2015-04-02 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
US20150118264A1 (en) 2012-01-31 2015-04-30 Curevac Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
US20150118183A1 (en) 2012-01-31 2015-04-30 Curevac Gmbh Negatively charged nucleic acid comprising complexes for immunostimulation
US20150165006A1 (en) 2012-02-15 2015-06-18 Cure GmbH Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded allergenic antigen or an autoimmune self-antigen
US20150184195A1 (en) 2012-03-27 2015-07-02 Curevac Gmbh Artificial nucleic acid molecules
US20150218554A1 (en) 2012-03-27 2015-08-06 Curevac Gmbh Artificial nucleic acid molecules for improved protein or peptide expression
US20150306249A1 (en) 2012-05-25 2015-10-29 Curevac Gmbh Reversible immobilization and/or controlled release of nucleic acid containing nanoparticles by (biodegradable) polymer coatings
US20150320847A1 (en) 2012-02-15 2015-11-12 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded tumour antigen
US20160024493A1 (en) 2013-03-15 2016-01-28 Adaptive Biotechnologies Corporation Uniquely tagged rearranged adaptive immune receptor genes in a complex gene set
US20160046949A1 (en) * 2013-03-14 2016-02-18 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US20160130345A1 (en) 2013-02-22 2016-05-12 Curevac Ag Combination of vaccination and inhibition of the pd-1 pathway
US20160166668A1 (en) 2013-08-21 2016-06-16 Curevac Ag Composition and vaccine for treating prostate cancer
US20160168227A1 (en) 2013-08-21 2016-06-16 Curevac Ag Composition and vaccine for treating lung cancer
US20160166678A1 (en) 2013-08-21 2016-06-16 Curevac Ag Combination vaccine
US20160166710A1 (en) 2013-08-21 2016-06-16 Curevac Ag Method for increasing expression of rna-encoded proteins
US20160168207A1 (en) 2013-08-21 2016-06-16 Curevac Ag Respiratory syncytial virus (rsv) vaccine
US20160166711A1 (en) 2013-08-21 2016-06-16 Curevac Ag Rabies vaccine
US20160235864A1 (en) 2013-11-01 2016-08-18 Curevac Ag Modified rna with decreased immunostimulatory properties
US20160304938A1 (en) 2013-12-30 2016-10-20 Curevac Ag Methods for rna analysis
US20160304883A1 (en) 2013-12-30 2016-10-20 Curevac Ag Artificial nucleic acid molecules
US20160326575A1 (en) 2015-05-08 2016-11-10 Curevac Ag Method for producing rna
US20160331844A1 (en) 2015-04-22 2016-11-17 Curevac Ag Rna containing composition for treatment of tumor diseases
WO2016184576A2 (fr) 2015-05-20 2016-11-24 Curevac Ag Composition de poudre sèche comprenant de l'arn à chaîne longue
WO2017001058A1 (fr) 2015-07-01 2017-01-05 Curevac Ag Procédé d'analyse d'une molécule d'arn
US20170014496A1 (en) 2014-03-12 2017-01-19 Curevac Ag Combination of vaccination and ox40 agonists
US20170029847A1 (en) 2013-12-30 2017-02-02 Curevac Ag Artificial nucleic acid molecules
WO2017021546A1 (fr) 2015-08-05 2017-02-09 Curevac Ag Vaccin d'arnm épidermique
WO2017025447A1 (fr) 2015-08-10 2017-02-16 Curevac Ag Procédé d'augmentation de la réplication d'une molécule d'adn circulaire
WO2017064146A1 (fr) 2015-10-12 2017-04-20 Curevac Ag Procédé automatisé d'isolement, de sélection et/ou de détection de micro-organismes ou de cellules présents dans une solution
US20170114378A1 (en) 2014-06-10 2017-04-27 Curevac Ag Methods and means for enhancing rna production
WO2017081110A1 (fr) 2015-11-09 2017-05-18 Curevac Ag Vaccins contre les rotavirus
WO2017098468A1 (fr) 2015-12-09 2017-06-15 Novartis Ag Analyse sans usage de marqueurs de l'efficacité d'un coiffage de l'arn à l'aide de la ribonucléase h, de sondes et de la chromatographie en phase liquide couplée à la spectrométrie de masse
US20170183700A1 (en) 2014-05-30 2017-06-29 Sc World, Inc. Method for Amplifying TCR cDNA
WO2017109134A1 (fr) 2015-12-22 2017-06-29 Curevac Ag Procédé de production de compositions de molécules d'arn
WO2017137095A1 (fr) 2016-02-12 2017-08-17 Curevac Ag Procédé d'analyse d'arn
WO2017140905A1 (fr) 2016-02-17 2017-08-24 Curevac Ag Vaccin contre le virus zika
WO2017140345A1 (fr) 2016-02-15 2017-08-24 Curevac Ag Procédé permet d'analyser des sous-produits de transcription in vitro d'arn
US20170252430A1 (en) 2014-04-01 2017-09-07 Curevac Ag Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
WO2017149139A1 (fr) 2016-03-03 2017-09-08 Curevac Ag Analyse d'arn par hydrolyse totale
WO2017162297A1 (fr) 2016-03-24 2017-09-28 Curevac Ag Pyrophosphatase inorganique immobilisée (ppase)
WO2017182634A1 (fr) 2016-04-22 2017-10-26 Curevac Ag Arn codant pour un antigène tumoral
WO2017186928A1 (fr) 2016-04-29 2017-11-02 Curevac Ag Arn codant pour un anticorps
WO2017191274A2 (fr) 2016-05-04 2017-11-09 Curevac Ag Arn codant pour une protéine thérapeutique
WO2017191264A1 (fr) 2016-05-04 2017-11-09 Curevac Ag Molécules d'acide nucléique et leurs utilisations
WO2017191258A1 (fr) 2016-05-04 2017-11-09 Curevac Ag Vaccins contre l'arnm de la grippe
US20170326225A1 (en) 2014-12-16 2017-11-16 Curevac Ag Ebolavirus and marburgvirus vaccines
WO2017203008A1 (fr) 2016-05-25 2017-11-30 Curevac Ag Nouveaux biomarqueurs
WO2017212007A1 (fr) 2016-06-09 2017-12-14 Curevac Ag Supports cationiques destinés à l'administration d'acides nucléiques
WO2017212006A1 (fr) 2016-06-09 2017-12-14 Curevac Ag Supports hybrides pour charge d'acide nucléique
WO2017212009A1 (fr) 2016-06-09 2017-12-14 Curevac Ag Supports hybrides pour cargo d'acides nucléiques
WO2017212008A1 (fr) 2016-06-09 2017-12-14 Curevac Ag Supports hybrides pour charge d'acide nucléique
US20180044687A1 (en) 2014-12-12 2018-02-15 Curevac Ag Artificial nucleic acid molecules for improved protein expression
US20180066325A1 (en) 2015-05-18 2018-03-08 Calimmune, Inc. Methods for discriminating between hiv-1 and lentiviral vectors
WO2018078053A1 (fr) 2016-10-26 2018-05-03 Curevac Ag Vaccins à arnm à nanoparticules lipidiques
US20180126003A1 (en) 2016-05-04 2018-05-10 Curevac Ag New targets for rna therapeutics
US20180125952A1 (en) 2015-05-15 2018-05-10 Curevac Ag PRIME-BOOST REGIMENS INVOLVING ADMINISTRATION OF AT LEAST ONE mRNA CONSTRUCT
US20180142275A1 (en) 2015-04-30 2018-05-24 Curevac Ag Immobilized poly(n)polymerase
US20180148727A1 (en) 2014-12-30 2018-05-31 Curevac Ag Artificial nucleic acid molecules
WO2018096179A1 (fr) 2016-11-28 2018-05-31 Curevac Ag Procédé de purification d'arn
US20180147146A1 (en) 2015-05-20 2018-05-31 Curevac Ag Dry powder composition comprising long-chain rna
WO2018104540A1 (fr) 2016-12-08 2018-06-14 Curevac Ag Arn pour la cicatrisation des plaies
US20180201967A1 (en) 2015-07-13 2018-07-19 Curevac Ag Method of producing rna from circular dna and corresponding template dna
US20180208957A1 (en) 2015-04-30 2018-07-26 Curevac Ag Method for in vitro transcription using an immobilized restriction enzyme
US20180214537A1 (en) 2015-04-13 2018-08-02 Curevac Ag Method for producing rna compositions
US20180237817A1 (en) 2015-05-29 2018-08-23 Curevac Ag Method for adding cap structures to rna using immobilized enzymes
US20180237786A1 (en) 2015-08-28 2018-08-23 Curevac Ag Artificial nucleic acid molecules
US20180243219A1 (en) 2015-04-17 2018-08-30 Curevac Ag Lyophilization of rna
WO2018167320A1 (fr) 2017-03-17 2018-09-20 Curevac Ag Vaccin à arn et inhibiteurs de points de contrôle immunitaires pour une thérapie anticancéreuse combinée
WO2018172556A1 (fr) 2017-03-24 2018-09-27 Curevac Ag Acides nucléiques codant pour des protéines associées à crispr et leurs utilisations
US20180296663A1 (en) 2015-06-17 2018-10-18 Curevac Ag Vaccine composition
US20180298372A1 (en) 2015-05-29 2018-10-18 Curevac Ag A method for producing and purifying rna, comprising at least one step of tangential flow filtration
US20180312545A1 (en) 2015-11-09 2018-11-01 Curevac Ag Optimized nucleic acid molecules
WO2018211038A1 (fr) 2017-05-17 2018-11-22 Curevac Ag Procédé de détermination d'au moins un paramètre de qualité d'un échantillon d'arn
US20180371392A1 (en) 2015-12-21 2018-12-27 Curevac Ag Inlay for a culture plate and corresponding method for preparing a culture plate system with such inlay
WO2019008001A1 (fr) 2017-07-04 2019-01-10 Curevac Ag Nouvelles molécules d'acide nucléique
US20190010485A1 (en) 2015-12-23 2019-01-10 Curevac Ag Method of rna in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
US20190024096A1 (en) 2015-08-07 2019-01-24 Curevac Ag Process for the in vivo production of rna in a host cell
US20190185859A1 (en) 2016-08-19 2019-06-20 Curevac Ag Rna for cancer therapy
US20190343933A1 (en) 2016-12-08 2019-11-14 Curevac Ag Rna for treatment or prophylaxis of a liver disease
US20190351047A1 (en) 2016-12-23 2019-11-21 Curevac Ag Henipavirus vaccine
US20190351048A1 (en) 2016-12-23 2019-11-21 Curevac Ag Mers coronavirus vaccine
US20190351044A1 (en) 2016-12-23 2019-11-21 Curevac Ag Lassa virus vaccine
US20200392572A1 (en) 2017-12-21 2020-12-17 Curevac Ag Linear double stranded dna coupled to a single support or a tag and methods for producing said linear double stranded dna
US20210030864A1 (en) 2017-08-22 2021-02-04 Curevac Ag Bunyavirales vaccine
US20210069315A1 (en) 2017-12-13 2021-03-11 Curevac Ag Flavivirus vaccine
US20210170017A1 (en) 2018-04-17 2021-06-10 Curevac Ag Novel rsv rna molecules and compositions for vaccination
WO2021123332A1 (fr) 2019-12-20 2021-06-24 Curevac Ag Nanoparticules lipidiques pour l'administration d'acides nucléiques
US20210261897A1 (en) 2018-06-28 2021-08-26 Curevac Ag Bioreactor for rna in vitro transcription
US20210260178A1 (en) 2018-06-27 2021-08-26 Curevac Ag Novel lassa virus rna molecules and compositions for vaccination
US20210361761A1 (en) 2018-04-05 2021-11-25 Curevac Ag Novel yellow fever nucleic acid molecules for vaccination
US20210379181A1 (en) 2020-02-04 2021-12-09 Curevac Ag Coronavirus vaccine
WO2021254593A1 (fr) 2020-06-15 2021-12-23 Curevac Ag Analyse de mélanges d'acides nucléiques
WO2022023559A1 (fr) 2020-07-31 2022-02-03 Curevac Ag Mélanges d'anticorps codés par des acides nucléiques
US20220040281A1 (en) 2018-12-21 2022-02-10 Curevac Ag Rna for malaria vaccines
WO2022043551A2 (fr) 2020-08-31 2022-03-03 Curevac Ag Vaccins contre le coronavirus à base d'acides nucléiques multivalents
US20220073962A1 (en) 2018-12-21 2022-03-10 Curevac Ag Methods for rna analysis
WO2022049093A1 (fr) 2020-09-01 2022-03-10 CureVac RNA Printer GmbH Dispositif de fabrication d'un produit pharmaceutique
US20220133908A1 (en) 2019-02-08 2022-05-05 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophthalmic diseases
US20220144877A1 (en) 2017-11-08 2022-05-12 Curevac Ag Rna sequence adaptation
US20220211838A1 (en) 2020-05-29 2022-07-07 Curevac Ag Nucleic acid based combination vaccines
US20220233568A1 (en) 2017-10-19 2022-07-28 Curevac Ag Novel artificial nucleic acid molecules
US20220296628A1 (en) 2019-08-14 2022-09-22 Curevac Ag Rna combinations and compositions with decreased immunostimulatory properties
US20220313813A1 (en) 2019-06-18 2022-10-06 Curevac Ag Rotavirus mrna vaccine
US20220340641A1 (en) 2018-12-14 2022-10-27 Curevac Ag Kras variant mrna molecules

Patent Citations (181)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050032730A1 (en) 2001-06-05 2005-02-10 Florian Von Der Mulbe Pharmaceutical composition containing a stabilised mRNA optimised for translation in its coding regions
WO2002098443A2 (fr) 2001-06-05 2002-12-12 Curevac Gmbh Composition pharmaceutique contenant un arnm stabilise et optimise pour la traduction dans ses regions codantes
US20060275747A1 (en) 2001-12-07 2006-12-07 Hardy Stephen F Endogenous retrovirus up-regulated in prostate cancer
US20050059624A1 (en) 2001-12-19 2005-03-17 Ingmar Hoerr Application of mRNA for use as a therapeutic against tumour diseases
US7074596B2 (en) 2002-03-25 2006-07-11 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Synthesis and use of anti-reverse mRNA cap analogues
US20050250723A1 (en) 2002-07-03 2005-11-10 Ingmar Hoerr Immunostimulation by chemically modified RNA
US20060188490A1 (en) 2003-08-05 2006-08-24 Ingmar Hoerr Transfection of blood cells with mRNA for immune stimulation and gene therapy
EP1518928A1 (fr) 2003-09-26 2005-03-30 Fundacion Pablo Cassara Oligonucléotides à activité antiandrogène pour le traitement des désordres cutanés associés aux androgènes. Compositions pharmaceutiques et leur utilisation
US20080025944A1 (en) 2004-09-02 2008-01-31 Cure Vac Gmbh Combination Therapy for Immunostimulation
EP2742951A2 (fr) 2004-09-14 2014-06-18 Argos Therapeutics, Inc. Amplification de souches indépendantes d'polynucleotides de VIH
US20080267873A1 (en) 2005-05-19 2008-10-30 Curevac Gmbh Injection Solution for Rna
WO2008016473A2 (fr) 2006-07-28 2008-02-07 Applera Corporation ANALOGUES DE COIFFES DE NUCLÉOTIDE D'ARNm
US20090324584A1 (en) 2006-07-31 2009-12-31 Curevac Gmbh Nucleic Acid of Formula (I): GIXmGn, or (II): CIXmCn, in Particular as an Immune-Stimulating Agent/Adjuvant
WO2008077592A1 (fr) 2006-12-22 2008-07-03 Curevac Gmbh Procédé de purification d'arn à l'échelle préparative par hplc
US20100048883A1 (en) 2006-12-22 2010-02-25 Thomas Ketterer Method for purifying rna on a preparative scale by means of hplc
US20100189729A1 (en) 2007-01-09 2010-07-29 Curvac Gmbh Rna-coded antibody
WO2008157688A2 (fr) 2007-06-19 2008-12-24 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Synthèse et utilisation d'analogues de phosphorothioate anti-inverse de la coiffe d'arn messager
US20100203076A1 (en) 2007-09-04 2010-08-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
US20100305196A1 (en) 2007-10-09 2010-12-02 Cure Vac Gmbh COMPOSITION FOR TREATING PROSTATE CANCER (PCa)
US20100291156A1 (en) 2007-10-09 2010-11-18 Marijke Barner Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US20120021043A1 (en) 2008-01-31 2012-01-26 CureVac GmbHÛ Nucleic Acids Comprising formula (NuGlXmGnNv)a and Derivatives Thereof as Immunostimulating Agent/Adjuvant
WO2009149253A2 (fr) 2008-06-06 2009-12-10 Uniwersytet Warszawski Analogues d'arnm cap
US20110250225A1 (en) 2008-09-30 2011-10-13 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
WO2010093820A2 (fr) 2009-02-11 2010-08-19 Orion Genomics Llc Combinaisons de polymorphismes destinées à déterminer l'expression spécifique des allèles d'igf2
WO2010123501A1 (fr) 2009-04-22 2010-10-28 Massachusetts Institute Of Technology Suppression immunitaire innée permettant la distribution répétée de longues molécules d'arn
WO2011015347A1 (fr) 2009-08-05 2011-02-10 Biontech Ag Composition vaccinale contenant de l'arn dont la coiffe en 5' est modifiée
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
US20120258046A1 (en) 2009-12-09 2012-10-11 Thorsten Mutzke Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
US20130142818A1 (en) 2010-07-30 2013-06-06 Curevac Gmbh Complexation of nucleic acids with disulfide-crosslinked cationic components for transfection and immunostimulation
WO2012019630A1 (fr) 2010-08-13 2012-02-16 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'une protéine codée
WO2012019780A1 (fr) 2010-08-13 2012-02-16 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'une protéine codée
US20130129754A1 (en) 2010-08-13 2013-05-23 Andreas Thess Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
US20130280283A1 (en) 2010-12-29 2013-10-24 Curevac Gmbh Combination of vaccination and inhibition of mhc class i restricted antigen presentation
WO2012104360A1 (fr) 2011-02-03 2012-08-09 University Of Veterinary Medicine Vienna Test rt-pcr pour la détection du virus de l'hépatite e aviaire
US20130259879A1 (en) 2011-02-21 2013-10-03 Curevac Gmbh Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
US20130336998A1 (en) 2011-03-02 2013-12-19 Curevac Gmbh Vaccination in newborns and infants
US20130295043A1 (en) 2011-03-02 2013-11-07 Curevac Gmbh Vaccination in elderly patients
WO2012135805A2 (fr) 2011-03-31 2012-10-04 modeRNA Therapeutics Administration et formulation d'acides nucléiques génétiquement modifiés
WO2013049231A1 (fr) 2011-09-28 2013-04-04 Htg Molecular Diagnostics, Inc. Procédés de co-détection d'arnm et d'un petit arn non codant
WO2013052523A1 (fr) 2011-10-03 2013-04-11 modeRNA Therapeutics Nucléosides, nucléotides et acides nucléiques modifiés, et leurs utilisations
WO2013059475A1 (fr) 2011-10-18 2013-04-25 Life Technologies Corporation Analogues de coiffes à dérivation alcynyle, préparation et utilisations associées
US20150118183A1 (en) 2012-01-31 2015-04-30 Curevac Gmbh Negatively charged nucleic acid comprising complexes for immunostimulation
US20150118264A1 (en) 2012-01-31 2015-04-30 Curevac Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
US20150037326A1 (en) 2012-01-31 2015-02-05 Bayer Innovation Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and an antigen
US20150320847A1 (en) 2012-02-15 2015-11-12 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded tumour antigen
US20150093413A1 (en) 2012-02-15 2015-04-02 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
US20150165006A1 (en) 2012-02-15 2015-06-18 Cure GmbH Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded allergenic antigen or an autoimmune self-antigen
US20150057340A1 (en) 2012-02-15 2015-02-26 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
US20150050302A1 (en) 2012-03-27 2015-02-19 Curevac Gmbh Artificial nucleic acid molecules comprising a 5'top utr
US20150184195A1 (en) 2012-03-27 2015-07-02 Curevac Gmbh Artificial nucleic acid molecules
US20150218554A1 (en) 2012-03-27 2015-08-06 Curevac Gmbh Artificial nucleic acid molecules for improved protein or peptide expression
WO2013143700A2 (fr) 2012-03-27 2013-10-03 Curevac Gmbh Molécules d'acide nucléique artificielles comprenant une 5'top utr
US20150306249A1 (en) 2012-05-25 2015-10-29 Curevac Gmbh Reversible immobilization and/or controlled release of nucleic acid containing nanoparticles by (biodegradable) polymer coatings
WO2014005038A1 (fr) 2012-06-29 2014-01-03 Htg Molecular Diagnostics, Inc. Procédés de protection d'une nucléase pour la détection de variants nucléotidiques
US20160130345A1 (en) 2013-02-22 2016-05-12 Curevac Ag Combination of vaccination and inhibition of the pd-1 pathway
WO2014152659A1 (fr) 2013-03-14 2014-09-25 Shire Human Genetic Therapies, Inc. Évaluation quantitative pour l'efficacité de coiffage de l'arn messager
US20160046949A1 (en) * 2013-03-14 2016-02-18 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US20160024493A1 (en) 2013-03-15 2016-01-28 Adaptive Biotechnologies Corporation Uniquely tagged rearranged adaptive immune receptor genes in a complex gene set
WO2015001416A1 (fr) 2013-07-05 2015-01-08 Tata Consultancy Services Limited Regroupement de données multidimensionnelles
US20160168227A1 (en) 2013-08-21 2016-06-16 Curevac Ag Composition and vaccine for treating lung cancer
US20160166678A1 (en) 2013-08-21 2016-06-16 Curevac Ag Combination vaccine
US20160166710A1 (en) 2013-08-21 2016-06-16 Curevac Ag Method for increasing expression of rna-encoded proteins
US20160168207A1 (en) 2013-08-21 2016-06-16 Curevac Ag Respiratory syncytial virus (rsv) vaccine
US20160166711A1 (en) 2013-08-21 2016-06-16 Curevac Ag Rabies vaccine
US20160166668A1 (en) 2013-08-21 2016-06-16 Curevac Ag Composition and vaccine for treating prostate cancer
US20160235864A1 (en) 2013-11-01 2016-08-18 Curevac Ag Modified rna with decreased immunostimulatory properties
US20170029847A1 (en) 2013-12-30 2017-02-02 Curevac Ag Artificial nucleic acid molecules
US20160304938A1 (en) 2013-12-30 2016-10-20 Curevac Ag Methods for rna analysis
US20160304883A1 (en) 2013-12-30 2016-10-20 Curevac Ag Artificial nucleic acid molecules
US20170014496A1 (en) 2014-03-12 2017-01-19 Curevac Ag Combination of vaccination and ox40 agonists
US20170252430A1 (en) 2014-04-01 2017-09-07 Curevac Ag Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
US20170183700A1 (en) 2014-05-30 2017-06-29 Sc World, Inc. Method for Amplifying TCR cDNA
US20170114378A1 (en) 2014-06-10 2017-04-27 Curevac Ag Methods and means for enhancing rna production
US20180044687A1 (en) 2014-12-12 2018-02-15 Curevac Ag Artificial nucleic acid molecules for improved protein expression
US20170326225A1 (en) 2014-12-16 2017-11-16 Curevac Ag Ebolavirus and marburgvirus vaccines
US20180148727A1 (en) 2014-12-30 2018-05-31 Curevac Ag Artificial nucleic acid molecules
US20180214537A1 (en) 2015-04-13 2018-08-02 Curevac Ag Method for producing rna compositions
US20180243219A1 (en) 2015-04-17 2018-08-30 Curevac Ag Lyophilization of rna
US20160331844A1 (en) 2015-04-22 2016-11-17 Curevac Ag Rna containing composition for treatment of tumor diseases
US20180142275A1 (en) 2015-04-30 2018-05-24 Curevac Ag Immobilized poly(n)polymerase
US20180208957A1 (en) 2015-04-30 2018-07-26 Curevac Ag Method for in vitro transcription using an immobilized restriction enzyme
US20160326575A1 (en) 2015-05-08 2016-11-10 Curevac Ag Method for producing rna
US20180125952A1 (en) 2015-05-15 2018-05-10 Curevac Ag PRIME-BOOST REGIMENS INVOLVING ADMINISTRATION OF AT LEAST ONE mRNA CONSTRUCT
US20180066325A1 (en) 2015-05-18 2018-03-08 Calimmune, Inc. Methods for discriminating between hiv-1 and lentiviral vectors
US20180147146A1 (en) 2015-05-20 2018-05-31 Curevac Ag Dry powder composition comprising long-chain rna
US20190133950A1 (en) 2015-05-20 2019-05-09 Curevac Ag Dry powder composition comprising long-chain rna
WO2016184576A2 (fr) 2015-05-20 2016-11-24 Curevac Ag Composition de poudre sèche comprenant de l'arn à chaîne longue
US20180237817A1 (en) 2015-05-29 2018-08-23 Curevac Ag Method for adding cap structures to rna using immobilized enzymes
US20180298372A1 (en) 2015-05-29 2018-10-18 Curevac Ag A method for producing and purifying rna, comprising at least one step of tangential flow filtration
US20180296663A1 (en) 2015-06-17 2018-10-18 Curevac Ag Vaccine composition
US20190017100A1 (en) 2015-07-01 2019-01-17 Curevac Ag Method for analysis of an rna molecule
WO2017001058A1 (fr) 2015-07-01 2017-01-05 Curevac Ag Procédé d'analyse d'une molécule d'arn
US20180201967A1 (en) 2015-07-13 2018-07-19 Curevac Ag Method of producing rna from circular dna and corresponding template dna
WO2017021546A1 (fr) 2015-08-05 2017-02-09 Curevac Ag Vaccin d'arnm épidermique
US20200085852A1 (en) 2015-08-05 2020-03-19 Curevac Ag Epidermal mrna vaccine
US20190024096A1 (en) 2015-08-07 2019-01-24 Curevac Ag Process for the in vivo production of rna in a host cell
WO2017025447A1 (fr) 2015-08-10 2017-02-16 Curevac Ag Procédé d'augmentation de la réplication d'une molécule d'adn circulaire
US20190225971A1 (en) 2015-08-10 2019-07-25 Curevac Ag Method of increasing the replication of a circular dna molecule
US20180237786A1 (en) 2015-08-28 2018-08-23 Curevac Ag Artificial nucleic acid molecules
WO2017064146A1 (fr) 2015-10-12 2017-04-20 Curevac Ag Procédé automatisé d'isolement, de sélection et/ou de détection de micro-organismes ou de cellules présents dans une solution
US20190249219A1 (en) 2015-10-12 2019-08-15 Curevac Ag Automated method for isolation, selection and/or detection of microorganisms or cells comprised in a solution
US20190160164A1 (en) 2015-11-09 2019-05-30 Curevac Ag Rotavirus vaccines
US20180312545A1 (en) 2015-11-09 2018-11-01 Curevac Ag Optimized nucleic acid molecules
WO2017081110A1 (fr) 2015-11-09 2017-05-18 Curevac Ag Vaccins contre les rotavirus
WO2017098468A1 (fr) 2015-12-09 2017-06-15 Novartis Ag Analyse sans usage de marqueurs de l'efficacité d'un coiffage de l'arn à l'aide de la ribonucléase h, de sondes et de la chromatographie en phase liquide couplée à la spectrométrie de masse
US20180371392A1 (en) 2015-12-21 2018-12-27 Curevac Ag Inlay for a culture plate and corresponding method for preparing a culture plate system with such inlay
US20190083602A1 (en) 2015-12-22 2019-03-21 Curevac Ag Method for producing rna molecule compositions
WO2017109134A1 (fr) 2015-12-22 2017-06-29 Curevac Ag Procédé de production de compositions de molécules d'arn
US20190010485A1 (en) 2015-12-23 2019-01-10 Curevac Ag Method of rna in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
US20210180106A1 (en) 2016-02-12 2021-06-17 Curevac Ag Method for analyzing rna
WO2017137095A1 (fr) 2016-02-12 2017-08-17 Curevac Ag Procédé d'analyse d'arn
US20190049414A1 (en) 2016-02-15 2019-02-14 Curevac Ag Method for analyzing by-products of rna in vitro transcription
WO2017140345A1 (fr) 2016-02-15 2017-08-24 Curevac Ag Procédé permet d'analyser des sous-produits de transcription in vitro d'arn
US20210205434A1 (en) 2016-02-17 2021-07-08 Curevac Ag Zika virus vaccine
WO2017140905A1 (fr) 2016-02-17 2017-08-24 Curevac Ag Vaccin contre le virus zika
US20190100784A1 (en) 2016-03-03 2019-04-04 Curevac Ag Rna analysis by total hydrolysis
WO2017149139A1 (fr) 2016-03-03 2017-09-08 Curevac Ag Analyse d'arn par hydrolyse totale
WO2017162297A1 (fr) 2016-03-24 2017-09-28 Curevac Ag Pyrophosphatase inorganique immobilisée (ppase)
US20190177714A1 (en) 2016-03-24 2019-06-13 Curevac Ag Immobilized inorganic pyrophosphatase (ppase)
WO2017182634A1 (fr) 2016-04-22 2017-10-26 Curevac Ag Arn codant pour un antigène tumoral
US20190343942A1 (en) 2016-04-22 2019-11-14 Curevac Ag Rna encoding a tumor antigen
WO2017186928A1 (fr) 2016-04-29 2017-11-02 Curevac Ag Arn codant pour un anticorps
US20200023076A1 (en) 2016-04-29 2020-01-23 Curevac Ag Rna encoding an antibody
WO2017191274A2 (fr) 2016-05-04 2017-11-09 Curevac Ag Arn codant pour une protéine thérapeutique
US20180126003A1 (en) 2016-05-04 2018-05-10 Curevac Ag New targets for rna therapeutics
WO2017191264A1 (fr) 2016-05-04 2017-11-09 Curevac Ag Molécules d'acide nucléique et leurs utilisations
WO2017191258A1 (fr) 2016-05-04 2017-11-09 Curevac Ag Vaccins contre l'arnm de la grippe
US20210162037A1 (en) 2016-05-04 2021-06-03 Curevac Ag Influenza mrna vaccines
US20190241633A1 (en) 2016-05-04 2019-08-08 Curevac Ag Rna encoding a therapeutic protein
US20190125857A1 (en) 2016-05-04 2019-05-02 Curevac Ag Nucleic acid molecules and uses thereof
WO2017203008A1 (fr) 2016-05-25 2017-11-30 Curevac Ag Nouveaux biomarqueurs
US20190194760A1 (en) 2016-05-25 2019-06-27 Curevac Ag Novel biomarkers
US20200179526A1 (en) 2016-06-09 2020-06-11 Curevac Ag Hybrid carriers for nucleic acid cargo
US20190381180A1 (en) 2016-06-09 2019-12-19 Curevac Ag Hybrid carriers for nucleic acid cargo
WO2017212007A1 (fr) 2016-06-09 2017-12-14 Curevac Ag Supports cationiques destinés à l'administration d'acides nucléiques
WO2017212008A1 (fr) 2016-06-09 2017-12-14 Curevac Ag Supports hybrides pour charge d'acide nucléique
WO2017212006A1 (fr) 2016-06-09 2017-12-14 Curevac Ag Supports hybrides pour charge d'acide nucléique
WO2017212009A1 (fr) 2016-06-09 2017-12-14 Curevac Ag Supports hybrides pour cargo d'acides nucléiques
US20190336608A1 (en) 2016-06-09 2019-11-07 Curevac Ag Cationic carriers for nucleic acid delivery
US20190336611A1 (en) 2016-06-09 2019-11-07 Curevac Ag Hybrid carriers for nucleic acid cargo
US20190185859A1 (en) 2016-08-19 2019-06-20 Curevac Ag Rna for cancer therapy
US20200163878A1 (en) 2016-10-26 2020-05-28 Curevac Ag Lipid nanoparticle mrna vaccines
WO2018078053A1 (fr) 2016-10-26 2018-05-03 Curevac Ag Vaccins à arnm à nanoparticules lipidiques
US20200318097A1 (en) 2016-11-28 2020-10-08 Curevac Ag Method for purifying rna
WO2018096179A1 (fr) 2016-11-28 2018-05-31 Curevac Ag Procédé de purification d'arn
WO2018104540A1 (fr) 2016-12-08 2018-06-14 Curevac Ag Arn pour la cicatrisation des plaies
US20190343933A1 (en) 2016-12-08 2019-11-14 Curevac Ag Rna for treatment or prophylaxis of a liver disease
US20200149026A1 (en) 2016-12-08 2020-05-14 Curevac Ag Rnas for wound healing
US20190351044A1 (en) 2016-12-23 2019-11-21 Curevac Ag Lassa virus vaccine
US20190351048A1 (en) 2016-12-23 2019-11-21 Curevac Ag Mers coronavirus vaccine
US20190351047A1 (en) 2016-12-23 2019-11-21 Curevac Ag Henipavirus vaccine
US20200085944A1 (en) 2017-03-17 2020-03-19 Curevac Ag Rna vaccine and immune checkpoint inhibitors for combined anticancer therapy
WO2018167320A1 (fr) 2017-03-17 2018-09-20 Curevac Ag Vaccin à arn et inhibiteurs de points de contrôle immunitaires pour une thérapie anticancéreuse combinée
WO2018172556A1 (fr) 2017-03-24 2018-09-27 Curevac Ag Acides nucléiques codant pour des protéines associées à crispr et leurs utilisations
US20210403925A1 (en) 2017-03-24 2021-12-30 Curevac Ag Nucleic acids encoding crispr-associated proteins and uses thereof
US20230064106A1 (en) 2017-05-17 2023-03-02 Curevac Real Estate Gmbh Method for determining at least one quality parameter of an rna sample
WO2018211038A1 (fr) 2017-05-17 2018-11-22 Curevac Ag Procédé de détermination d'au moins un paramètre de qualité d'un échantillon d'arn
WO2019008001A1 (fr) 2017-07-04 2019-01-10 Curevac Ag Nouvelles molécules d'acide nucléique
US20190040378A1 (en) 2017-07-04 2019-02-07 Curevac Ag Novel nucleic acid molecules
US20210030864A1 (en) 2017-08-22 2021-02-04 Curevac Ag Bunyavirales vaccine
US20220233568A1 (en) 2017-10-19 2022-07-28 Curevac Ag Novel artificial nucleic acid molecules
US20220144877A1 (en) 2017-11-08 2022-05-12 Curevac Ag Rna sequence adaptation
US20210069315A1 (en) 2017-12-13 2021-03-11 Curevac Ag Flavivirus vaccine
US20200392572A1 (en) 2017-12-21 2020-12-17 Curevac Ag Linear double stranded dna coupled to a single support or a tag and methods for producing said linear double stranded dna
US20210361761A1 (en) 2018-04-05 2021-11-25 Curevac Ag Novel yellow fever nucleic acid molecules for vaccination
US20210170017A1 (en) 2018-04-17 2021-06-10 Curevac Ag Novel rsv rna molecules and compositions for vaccination
US20210260178A1 (en) 2018-06-27 2021-08-26 Curevac Ag Novel lassa virus rna molecules and compositions for vaccination
US20210261897A1 (en) 2018-06-28 2021-08-26 Curevac Ag Bioreactor for rna in vitro transcription
US20220340641A1 (en) 2018-12-14 2022-10-27 Curevac Ag Kras variant mrna molecules
US20220040281A1 (en) 2018-12-21 2022-02-10 Curevac Ag Rna for malaria vaccines
US20220073962A1 (en) 2018-12-21 2022-03-10 Curevac Ag Methods for rna analysis
US20220133908A1 (en) 2019-02-08 2022-05-05 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophthalmic diseases
US20220313813A1 (en) 2019-06-18 2022-10-06 Curevac Ag Rotavirus mrna vaccine
US20220296628A1 (en) 2019-08-14 2022-09-22 Curevac Ag Rna combinations and compositions with decreased immunostimulatory properties
WO2021123332A1 (fr) 2019-12-20 2021-06-24 Curevac Ag Nanoparticules lipidiques pour l'administration d'acides nucléiques
US20210379181A1 (en) 2020-02-04 2021-12-09 Curevac Ag Coronavirus vaccine
US20220211838A1 (en) 2020-05-29 2022-07-07 Curevac Ag Nucleic acid based combination vaccines
WO2021254593A1 (fr) 2020-06-15 2021-12-23 Curevac Ag Analyse de mélanges d'acides nucléiques
WO2022023559A1 (fr) 2020-07-31 2022-02-03 Curevac Ag Mélanges d'anticorps codés par des acides nucléiques
WO2022043551A2 (fr) 2020-08-31 2022-03-03 Curevac Ag Vaccins contre le coronavirus à base d'acides nucléiques multivalents
WO2022049093A1 (fr) 2020-09-01 2022-03-10 CureVac RNA Printer GmbH Dispositif de fabrication d'un produit pharmaceutique

Non-Patent Citations (37)

* Cited by examiner, † Cited by third party
Title
Andrus et al., "Base Composition Analysis of Nucleosides Using HPLC", In: Current Protocols in Nucleic Acid Chemistry, John Wiley & Sons, Inc., pp. 10.6.1-10.6.6, 2001.
Boyle et al., "Evaluation of the impact of single nucleotide polymorphisms and primer mismatches on quantitative PCR," BMC Biotechnology, 9:75, 2009.
Brunelle et al., "In vitro transcription from plasmid or PCR-amplified DNA", Methods Enzymol., 530:101-114, 2013.
Cohen et al., "High-performance capillary electrophoretic separation of bases, nucleosides, and oligonucleotides: retention manipulation via micellar solutions and metal additives", Anal. Chem., 59(7):1021-1027, 1987.
Danneberg et al., "Sequence-specific RNA cleavage by PNA conjugates of the metal-free artificial ribonuclease tris(2-aminobenzimidazole)," Beilstein Journal of Organic Chemistry, 11:493-498, 2015.
Desai and Shankar, "Single-strand-specific nucleases", FEMS Microbiol. Rev., 26:457-491, 2003.
Dogandzhiyski et al., "Studies on Tris(2-aminobenzimidazole)-PNA Based Artificial Nucleases: A Comparison of Two Analytical Techniques," Bioconjugate Chemistry, 26(12):2514-2519, 2015.
Dougherty et al., "Quantitative analysis of deadenylation-independent mRNA decay as a modified MBRACE assay," Methods Mol. Biol., 1125:353-371, 2014.
Eadie et al., "High-performance liquid chromatographic analysis of oligodeoxyribonucleotide base composition", Anal. Biochem., 165(2):442-447, 1987.
Fotin-Mleczek et al., "Highly potent mRNA based cancer vaccines represent an attractive platform for combination therapies supporting an improved therapeutic effect", J. Gene Med., 14(6):428-439, 2012.
Geall et al., "RNA: the new revolution in nucleic acid vaccines", Semin. Immunol., 25(2):152-159, 2013.
Heighway et al., "Coamplification in tumors of kras2, type 2 inositol 1,4,5 triphosphate receptor gene, and a novel human gene, krag", Genomics, 35(1):207-214, 1996.
International Search Report and Written Opinion issued in corresponding PCT Application No. PCT/EP2017/055048, dated May 16, 2017.
Johnsen et al., "Hydrophilic interaction chromatography of nucleoside triphosphates with temperature as a separation parameter", J. Chromatogr. A, 1218(35):5981-5986, 2011.
Kamiya et al., "Mutagenic properties of oxidized GTP and ATP in in vitro transcription-reverse transcription", Nucleic Acid Symposium Series, 50:99-100, 2006.
Kammerer et al., "MALDI-TOF MS analysis of urinary nucleosides", J. Am. Soc. Mass Spectrom., 16(6):940-947, 2005.
Kolpashchikov, "An elegant biosensor molecular beacon probe: challenges and recent solutions", Scientifica, 928783:1-17, 2012.
Kore et al., "Synthesis and biological validation of N7-(4-chlorophenoxyethyl) substituted dinucleotide cap analogs for mRNA translation", Bioorg. Med. Chem., 21(15):4570-4574, 2013.
Kuzuya et al., "Selective activation of two sites in RNA by acridine-bearing oligonucleotides for clipping of designated RNA fragments," Journal of the American Chemical Society, 126(5):1430-1436, 2004.
Li et al., "Development of an isocratic HPLC method for catechin quantification and its application to formulation studies", Fitoterapia, 83:1267-1274, 2012.
Li et al., "Ultrasensitive optical DNA biosensor based on surface immobilization of molecular beacon by a bridge structure", Anal. Sci., 17: 1149-1153, 2001.
Maxwell et al., "Assay of DNA-RNA hybrids by S 1 nuclease digestion and adsorption to DEAE-cellulose filters", Nucl. Acids. Res., 5(6):2033-2038, 1978.
Nakanishi et al., "Hair roots as an mRNA source for mutation analysis of Usher syndrome-causing genes," J Hum Genet., 55:701-703, 2010.
Nees et al., "Detection of RNA Modifications by HPLC Analysis and Competitive ELISA", In: Innate DNA and RNA Recognition, Anders and Migliorni (eds), 1169:3-14, 2014.
Nunomura et al., "Oxidative damage to RNA in aging and neurodegenerative disorders", Neurotox. Res., 22:231-248, 2012.
Shan et al., "Messenger RNA oxidation is an early event preceding cell death and causes reduced protein expression", FASEB J., 21:2753-2764, 2007.
Shimelis et al., "Nuclease P1 digestion/high-performance liquid chromatography, a practical method for DNA quantitation",J. Chromatog., 1117(2):132-136, 2006.
Stepinski et al., "Synthesis and properties of mRNAs containing the novel "anti-reverse" cap analogs 7-methyl(3′-O-methyl)GpppG and 7-methyl(3′-deoxy)GpppG", RNA, 7(10):1486-1495, 2001.
Su et al., "Quantitative analysis of ribonucleoside modifications in tRNA by HPLC-coupled mass spectrometry", Nature Protoc., 9(4):828-841, 2014.
Tanaka et al., "RNA oxidation catalyzed by cytochrome c leads to its depurination and cross-linking, which may facilitate cytochrome c release from mitochondria", Free Radic. Biol. Med., 53(4):854-862, 2012.
Theus and Liarakos, "A simple assay for determining the capping efficiencies of RNA polymerases used for in vitro transcription", BioTechniques, 9(5):610-615, 1990.
Thompson and Sommercorn, "Use of a multiple S1 nuclease protection assay to monitor changes in RNA levels for type 1 phosphatase and several proto-oncogenes in response to insulin", J. Biol. Chem., 267(9):5921-5926, 1992.
Vet and Marras, "Design and optimization of molecular beacon real-time polymerase chain reaction assays", In: Oligonucleotide Synthesis, Herdewijn, P. (ed.), 288:273-290, 2005.
Vet et al., "Multiplex detection of four pathogenic retroviruses using molecular beacons", Proc. Natl. Acad. Sci. USA, 96:6394-6399, 1999.
Wietstock, "DNA Composition Analysis by Nuclease Digestion and HPLC", J. Chem. Ed., 72(10):950, 1995.
Wong and Medrano, "Real-time PCR for mRNA quantitation", Biotechniques, 39(1):75-85, 2005.
Zuker, "Mfold web server for nucleic acid folding and hybridization prediction", Nucl. Acids Res., 31(13):3406-3415, 2003.

Also Published As

Publication number Publication date
US20190100784A1 (en) 2019-04-04
WO2017149139A1 (fr) 2017-09-08
EP3423595A1 (fr) 2019-01-09

Similar Documents

Publication Publication Date Title
US11920174B2 (en) RNA analysis by total hydrolysis and quantification of released nucleosides
US11268157B2 (en) Method for producing RNA
US20220119795A1 (en) Method of rna in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
US10501768B2 (en) Method of producing RNA from circular DNA and corresponding template DNA
US20190049414A1 (en) Method for analyzing by-products of rna in vitro transcription
US20230313268A1 (en) Methods for rna analysis
CN106661621B (zh) 用于增强rna产生的方法和工具
EP3303583B1 (fr) Procédé de production et de purification d'arn, comprenant au moins une étape de filtration à flux tangentiel
EP3540060A1 (fr) Procédés d'analyse d'arn
US11898186B1 (en) Compositions and methods for preparing capped mRNA
WO2018141371A1 (fr) Purification et/ou formulation d'arn

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: CUREVAC AG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:EBER, FABIAN JOHANNES;WOCHNER, ANIELA;ROOS, TILMANN;SIGNING DATES FROM 20190205 TO 20190206;REEL/FRAME:048452/0790

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

AS Assignment

Owner name: CUREVAC SE, GERMANY

Free format text: CHANGE OF NAME;ASSIGNOR:CUREVAC AG;REEL/FRAME:062683/0254

Effective date: 20220926

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

ZAAB Notice of allowance mailed

Free format text: ORIGINAL CODE: MN/=.

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE